You are on page 1of 19

Bone Research www.nature.

com/boneres

REVIEW ARTICLE OPEN

Bone remodeling: an operational process ensuring survival and


bone mechanical competence
1 1 1✉
Simona Bolamperti , Isabella Villa and Alessandro Rubinacci

Bone remodeling replaces old and damaged bone with new bone through a sequence of cellular events occurring on the same
surface without any change in bone shape. It was initially thought that the basic multicellular unit (BMU) responsible for bone
remodeling consists of osteoclasts and osteoblasts functioning through a hierarchical sequence of events organized into distinct
stages. However, recent discoveries have indicated that all bone cells participate in BMU formation by interacting both
simultaneously and at different differentiation stages with their progenitors, other cells, and bone matrix constituents. Therefore,
bone remodeling is currently considered a physiological outcome of continuous cellular operational processes optimized to
confer a survival advantage. Bone remodeling defines the primary activities that BMUs need to perform to renew successfully
bone structural units. Hence, this review summarizes the current understanding of bone remodeling and future research
directions with the aim of providing a clinically relevant biological background with which to identify targets for therapeutic
strategies in osteoporosis.

Bone Research (2022)10:48


1234567890();,:

; https://doi.org/10.1038/s41413-022-00219-8

INTRODUCTION BONE MODELING, REMODELING, AND THE MUTUAL


Despite the tremendous efforts of researchers studying bone REGULATION OF BONE RESORPTION AND FORMATION
remodeling for more than 50 years, the intrinsic spatial, In vertebrates, bone modeling and remodeling are essential
biomolecular, and mechanotransduction complexities in bone processes that are activated throughout life and are regulated by
remodeling continue to be debated. Initially, a two-stage distinct temporal cellular constituents that ensure functional bone
process involving two types of cellular machinery was adaptation and vertebrate survival. Bone modeling adapts bone
considered to be responsible for bone remodeling, in which shape to variable mechanical demands during growth and aging
bone formation by osteoblasts follows bone resorption by through cellular events that determine bone resorption and
osteoclasts to achieve net bone mass equilibrium upon formation on opposing cortical and cancellous surfaces. This
physiological maturity.1 However, recently identified cellular implies the existence of a modeling drift, which moves a bone
events, coordination pathways and anatomical structures have structural unit over time in the direction defined by growth
allowed a better understanding of the genesis, differentiation, patterns, and adjusts the bone mass distribution to the stresses
activity, crosstalk and death of the entire cell population and strains induced by locomotion and physical activity.3 By bone
involved in bone remodeling.2 These discoveries have shed remodeling, old or damaged bone is replaced with new bone
new light on the operational processes ensuring bone mass through a sequence of cellular events occurring on the same
renewal without bone mass loss under physiological condi- surface without any change in bone shape.4
tions. Bone remodeling is activated by local and systemic In the 1960s, Frost5 recognized that a forming osteon in
factors, supporting the concept that targeted remodeling is mammalian compact bone consists of a group of synchronous
activated by local factors and that stochastic remodeling is cells, suggesting “control mechanisms which are functionally and
activated by systemic factors, with these factors cooperating to temporally ordered, discontinuous and discrete”. Frost advanced
maintain mechanical competence and meet concurrent meta- the enlightening notion of a basic multicellular unit (BMU) as a
bolic demands. This new information implies that the determi- transient anatomic structure in bone remodeling and introduced
nants of the focal balance in bone mass after remodeling are the quantum concept of bone remodeling, which is analogous to
the integrated effects of both mechanical and metabolic quantum theory in physics. Quantum physics explains the property
environmental conditions. of matter at the smallest scale. It defines the behavior of the
This review provides a novel integrated picture of the minimum, discrete amount, i.e., the quantum, of any physical
operational bone remodeling processes by describing, updat- entity on the assumption that all phenomena in a submicroscopic
ing, and reexamining the current evidence and its biological system exhibit quantization.6 In an analogy, Frost intuitively
plausibility. In this review, bone remodeling is described defined a quantum of bone remodeling as a discrete change in
as a continuous flow of cellular signaling and connected bone mass resulting from the coordinated activity of an individual
events, not as a process comprising stages, as it has been BMU at an anatomically discrete locus.1 The quantum concept
historically presented. conceived by Frost and later extended by Parfitt7 has had

1
Osteoporosis and Bone and Mineral Metabolism Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132 Milano, Italy
Correspondence: Alessandro Rubinacci (rubinacci.alessandro@hsr.it)

Received: 2 August 2021 Revised: 2 May 2022 Accepted: 15 May 2022

© The Author(s) 2022


The operational process of bone remodeling
S Bolamperti et al.
2
Direction of BMU
propagation

Symmetry Nerve Cortical


line A similar
remodeling
process occurs
in cancellous
id
teo bone, but
Os within a canopy

The cutting Reversal Closing


cone zone zone

Reversal cell Osteoclast Osteocyte Pericyte

Osteoprogenitors Monocyte/ Active


macrophage osteoblast

Fig. 1 The cutting cone. The cutting cone originates in close proximity to neurovascular axial bundles and is generated by the propagation of
the basic multicellular unit (BMU). The cutting cone consists of a set of osteoclasts, followed by a set of osteoblasts, reversal cells and
secondary osteoclasts that cover the so-called reversal zone. At the end of the reversal zone, a set of osteocytes generate the closing zone. A
line of symmetry divides in half the representation of a complete BMU in the cortex moving toward the longitudinal axis of the long bone.
One-half of cortical BMU is similar to the BMU at the cancellous surface, although in cancellous bone, the BMU is separated from the marrow
by a specific cell structure called the canopy

profound implications for the understanding of all aspects of bone process couples the events necessary to remove and replace bone
pharmacology and physiopathology,8 particularly in the osteo- units under a hierarchy of time and space, independent of their
porotic context.9 A remarkable idea of the early sixties that is in focal balance; in fact, focal remodeling imbalance does not imply
line with the modern theory that quantum biological phenomena “uncoupling”. At each remodeling site, coupling indicates that the
can lead to evolutionary advantages.10 receptors on the osteoclast membrane as well as the regulatory
Frost recognized that the change in bone mass caused by the factors released by osteoclasts are coupled with the sequential
focal balance in each remodeling cycle of resorbed bone and recruitment and differentiation of osteoblast lineage cells toward
formed bone is not an outcome of isolated “working” cell packets the mature phenotype, laying down the bone matrix and inducing
but is derived from the interacting parts of the whole BMU, which mineralization. Coupling therefore implies that a commensurate
endures longer (9 months) than the lifespan of each single change in bone formation follows any pathology- or therapy-
component.4 This implies a continuous and ordered cell supply related modification of bone resorption.
that depends on the division frequency of each progenitor cell At each remodeling site, coupling is asynchronous: at any given
and the lifespan of each differentiated cell. A tightly maintained time, bone is being resorbed at some sites, while it is being formed
equilibrium between genesis and apoptosis is therefore critical for or is in the reversal phase from bone resorption to bone formation
a properly functioning BMU.11 Recent advances have added at other site. This implies that there is continuous focal transient
complexity to the original BMU description. The number of cells loss of bone that is fully reversed in balanced remodeling; as the
considered to constitute a BMU has expanded to include all bone number of activated remodeling sites increases, the transient loss
cells at all differentiation stages, interacting with their progenitors, of bone, which is defined as the “virtual space” of bone remodeling,
T cells and bone matrix components. The BMU includes, in increases. As coupling takes place in different locations at different
particular, a set of osteoclasts localized in the “cutting cone” times, it requires local regulatory factors to transfer information
followed by a set of cells, including reversal cells and osteoblasts, among cells and tissue locations according to bone remodeling
localized at the reversal zone; and a set of osteocytes localized in needs. Therefore, remodeling is defined as a dynamic physiological
the closing zone. These cell sets constitute a secondary osteon process executed through the coupled activity of osteoclasts and
around the neurovascular bundle wich is axially located with the osteoblasts belonging to a BMU. Conversely, bone turnover is the
connective matrix in the Haversian canal. A visualization of a outcome of bone remodeling that defines the amount of bone
complete BMU can be acquired only with a longitudinal section of removed and formed within a given volume in a given time and is
cortical bone, where Haversian canals run parallel to the sectional determined by the number of BMUs and by the focal balance
plane (Fig. 1). Notably, the 3D trabecular network prevents the within each BMU. As Parfitt said “Bone turnover refers to proportional
proper visualization of a complete BMU in cancellous bone.12 bone volume replacement per unit time, and is usually expressed as
In bone remodeling, osteoblast–osteoclast interactions are percent/year […]. If bone turnover is 10%/year, then the mean lifetime
necessary and must be coordinated in time and space to maintain of each moiety of bone is 10 years”.13
the focal bone remodeling balance; this balance refers to the net
amount of tissue resorbed and formed at each remodeling site
(i.e., focal point) to maintain the structural integrity of the tissue. OSTEOCYTES: TUNING, INITIATION AND TERMINATION OF
At maturity, the focal balance (neutral) in bone remodeling is BONE REMODELING
determined by equal bone aliquots, which are removed and Osteocytes are a relatively permanent phenotype of bone cells that
synthetized. In other periods in life or as a consequence of Frost estimated to have a lifespan of ~25 years1 and play a master
metabolic diseases, the focal balance can be negative (bone mass role in bone remodeling. Osteocytes represent more than 90% of
is lost) or positive (bone mass is gained). Focal coupling in bone bone cells in the adult skeleton14 with a average cell density highly
remodeling strictly refers to the transfer of the information within dependent on the specie, bone anatomical site, and subiect
the cell pool responsible for bone remodeling. This operational analyzed.15 Recent studies performed using new imaging

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
3
techniques have estimated that 19 000–28 500 cells per mm3 Ca2+-dependent contractions of the cell membrane favors the
populate the human skeleton, accounting for ~42 billion individual production and release of extracellular vesicles (EVs) containing
cells.16 Osteocytes consist of a stellate body connected by slender bone regulatory proteins.35 EVs are ubiquitous lipidic organelles
cell processes (50–100 processes per cell). The asymmetrical that mediate the intercellular transfer of information through their
“arborization” of dendrites polarizes toward bone surfaces, where cargo, which includes both proteins and nucleic acids.36 In
the dendrites contact osteoblasts undergoing growth or remodel- osteocytes, EVs can be observed in proximity of the osteocytic
ing or, during bone surface quiescence, bone lining cells. The term network.37 In response to mechanical stimuli and subsequent
“arborization” was originally introduced by Marotti and Palumbo,17 Ca2+ influx, osteocyte lines have been observed to release EVs
and it now refers to “dendrogenesis”, the asymmetric and containing RANKL, OPG, and sclerostin.35
asynchronous formation of osteocyte dendrites. Short radiating All the mechanical signals received by the osteocyte-bone
dendrites extend toward mineralized surface, and long radiating lining cell syncytium modulate cell apoptosis and survival as well
dendrites extend toward the vasculature during the progressive as the anabolic Wnt pathway in bone. Secreted Wnt protein
translocation of cell bodies farther from the vascular surface due to stimulates target cell via the β-catenin-mediated (canonical) and
the secretion of the osteoblastic lamina. Osteocytogenesis and β-catenin-independent (noncanonical) pathways. The canonical
dendrogenesis are discussed in a subsequent section (“The pathway has emerged as the predominant component of Wnt
osteoblast pool: recruitment, expansion and osteocytogenesis”). signaling in bone, positively affecting the entire osteoblast
An adult human skeleton includes 23 trillion osteocyte lineage.38 Wnt proteins bind to their receptors (Frizzled) and
connections with each other and with bone surface cells.16 coreceptors (LRP5/6) to promote the stabilization of β-catenin in
These connections form a 3D protoplasmic network that the cytoplasm, which translocates to the nucleus, where it induces
constitutes a matrix-integrated functional syncytium, which does the expression of osteogenesis-related genes.38,39 Many studies
not cross cement lines but does establish direct contact with the have demonstrated that β-catenin is required for bone formation
bone marrow, resides in low-oxygen microenvironment and and is activated during multiple stages of osteoblast differentia-
comprises multiple elements. During aging in both humans and tion to regulate both osteoblast and osteoclast.40–43 In fact, WNT–
rodents, this network deteriorates. Throughout aging, a large β-catenin signaling in osteoblasts and osteocytes indirectly
and linear reduction in dendrite number and cell body density represses osteoclast differentiation and bone resorption by
directly related to the deterioration of cortical parameters has stimulating the secretion of OPG.38
been observed.18 Since decreased osteocyte number accom- Mechanical signals activate the anabolic Wnt pathway in bone
panies reduced dendritic density, it has been suggested that through the suppression of the Wnt receptor antagonist sclerostin
dendrite loss might contribute to diminished osteocyte viability (SOST). Recently, Sato et al.20,44 showed that FFSS induces the
because a certain degree of locally and/or systemically triggered disruption of the FAK-integrin complex, thereby inhibiting histone
anabolic signaling through the osteocyte-lacunocanalicular deacetylase 4/5 (HDAC4/5), a negative regulator of SOST expres-
system is lost (see below).18 sion in osteocytes45 modulated by parathyroid hormone (PTH)
The osteocyte-bone lining cell syncytium displays gap junctions treatment.46 Osteocytes can also decrease SOST expression after
(connexins) that allow the transfer of information between cells. sensing decreases in oxygen levels.47
Connexin43 (Cx43) is the most abundant connexin in osteocytes, In vivo anatomical studies have suggested that the presence of
and global knockout of Cx43 expression is lethal at birth.19 apoptotic osteocytes at microdamaged sites correlates with the
Conditional knockout of Cx43 in osteoblasts and osteocytes in recruitment of osteoclasts at microcracks.48 In addition to their
mice led to various degrees of osteopenia depending on the fundamental role as mechanosensors, osteocytes appear to be
differentiation state in which the deletion was induced,20,21 spatially, temporally and mechanistically linked to bone remodel-
suggesting that functional Cx43 in osteoblasts and osteocytes is ing activation, particularly by regulating RANKL (as is discussed
essential for normal bone mass acquisition and maintenance. below). RANKL is routinely found as a membrane bound protein
The osteocyte-bone lining cell syncytium is endowed within the (mRANKL), but it can also be cleaved into a soluble form
lacunocanalicular network of cavities filled with bone extracellular (sRANKL)49 or delivered via EVs.35 RANKL binds the receptor
fluid (BEF). BEF has a different ionic composition from systemic activator of nuclear factor kappa-B (RANK) on osteoclasts to
extracellular fluid (SEF)22 and establishes an extensive contact induce osteoclastogenesis.50
surface (215 m2, which is 120-fold the size of the trabecular In 2011, Nakashima et al.51 showed that the conditional deletion
network) with the mineralized matrix to allow efficient, short-term of RANKL expression in osteocytes using a DMP1 promoter caused
mineral exchange with SEF.23–26 This specific physical environ- a severe osteopetrotic phenotype. This osteopetrotic phenotype
ment allows osteocytes to govern metabolic demands and was confirmed by Xiong et al.,52 who in addition found that RANKL
mechanotransduction for bone mass adaptation.27 The strain- mRNA, isolated from total bone of DMP1-cre+, RANKLloxp/loxP mice,
induced flow of BEF in the lacuno-canalicular network exerts a showed very little variation compared to that of wild type mice.
shear stress (fluid flow shear stress, FFSS) on osteocyte bodies, The concentration of circulating sRANKL was unaltered compared
which undergo dendrogenesis to activate several classes of to that in wild type animals. However, a 70% reduction in
mechano-sensors regulating specific gene expression patterns.28 osteoclast number in the cancellous bone of these transgenic
Osteocytes might sense FFSS through 1. “collagen hillocks”, mice highlighted the relevance of local RANKL production.52 As
which are collagen matrix projections in osteocyte canaliculi that the activation of Dmp1-Cre promoters is not exclusive to
directly link the matrix to osteocyte dendrites;29 2. β3 and β1 osteocytes,53 the conditional deletion of RANKL expression was
integrins, which participate to focal adhesion kinase (FAK) performed under the control of the more specific SOST promoter.
complex formation in osteocyte dendritic projections;30 3. primary This conditional deletion generated a phenotype resembling the
cilia, which have a flow-sensing function that leads to increased previous one, with a significant decrease in osteoclasts number in
osteoprotegerin (OPG)/receptor activator of nuclear factor kappa- cancellous bone.54 Taken together, these in vivo studies confirmed
B ligand (RANKL) ratio via a calcium-independent mechanism;31 4. that osteocytes produce RANKL and that osteocytic RANKL is
connexin43, a component of gap junctions that mediates the fundamental to sustaining bone remodeling.
transduction of mechanical signals;32 and 5. mechanosensitive ion Whether osteocyte RANKL is soluble, is transferred via vesicles
channels, such as those composed of Piezo1, which are highly or is membrane-bound is not fully clear. Bonewald et al.14 first
sensitive to osteocyte membrane tension.33 demonstrated that the MLO-Y4 osteocyte line expresses RANKL at
The deformation of the osteocyte cytoskeleton elicits Ca2+ the cell surface and at dendritic processes. MLO-Y4 cells and
influx signaling via the activation of TRPV4.34 The generation of primary murine osteocytes cocultured with bone marrow

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
4
Bone marrow Osteoclast precursor RANKL
RANK
Osteoclast

RANK RANK

Bcl-2
RANKL

P2Y2
RANKL

ATP ATP
P2Y2
ATP
P2X7
P2Y2

P2X7 Bcl-2 Panx1


ATP
Bcl-2 Panx1
Mic
roc By-stander
rac
k osteocyte

ATP
Apoptotic
P2Y2
P2X7 osteocyte
Extracellular Panx1
matrix

Fig. 2 The “find me” message from apoptotic osteocytes. Under fatigue failure, osteocytes undergo apoptosis, whereas the osteocytes
surrounding the apoptotic osteocytes, the “bystander osteocytes”, show upregulation of the antiapoptotic protein BcL-2, which protects them
from death. Apoptotic osteocytes activate the Panx1/P2XR pathway to induce the release of ATP into the extracellular compartment as a
specific “find me” signal. ATP binds P2Y2 receptors on bystander osteocytes, which in turn produce and release RANKL at the interface with
the bone marrow. RANKL promotes the recruitment of osteoclast progenitors and osteoclastogenesis

precursors supported osteoclastogenesis, but their conditioned soluble form is necessary to sustain bone remodeling under
media alone did not have a similar effect,55 suggesting that certain conditions.59 In support of this hypothesis, mice over-
cell–cell interactions between osteocytes and osteoclasts are expressing sRANKL only in the liver, and therefore also in blood
needed for osteoclastogenesis. In a different study, the condi- circulation, displayed a significant decrease in bone mass with an
tioned media of apoptotic MLO-Y4 cells increased osteoclast increase in the number of osteoclasts on cancellous bone surfaces
formation, osteoclast size and osteoclast precursor migration. during aging.60
Interestingly, in apoptotic MLO-Y4 cells, both RANKL mRNA and In vivo, osteocyte apoptosis is fundamental for initiating bone
protein expression were upregulated, suggesting that the condi- remodeling,61 independent of the stimulus causing osteocyte
tioned media of this cell culture might contain sRANKL.56 On the death such as estrogen loss,62 fatigue,63 or unloading.64 An in vivo
other hand, the conditioned media of a non apoptotic MLO-Y4 immunohistochemical analysis showed that the RANKL signal in
culture did not alter any of these aforementioned cell para- bone tissue was stronger in an area around 150–200 μm from the
meters.56 In another in vitro study, primary osteocytes were apopototc osteocytes.65 Interestingly, inhibiting apoptosis pre-
cocultured on a 3D collagen scaffold with osteoclast precursors vented the RANKL production by neighboring osteocytes,
separated by a porous membrane. Again, the osteocytes suggesting that “bone remodeling follows a common paradigm
supported osteoclast formation, but the efficiency of their effect for localized tissue repair”, with different response-induced factor
decreased with a decrease in the pore size of the membrane. released from apoptotic osteocytes near the damage site and
Confocal microscopy showed that large pore membranes allow “bystander” osteocytes.63 MLO-Y4 cell apoptosis generated ATP
osteocyte dendrites to touch osteoclast precursors through the release from cells via Pannexin 1 channels (PANX1) opening.66
pores, but this contact was not observed when small-pore The same phenotype was observed in mice treated with an
membranes were used.57 Therefore, the authors of this study inhibitor of the P2X7 receptor (P2X7R), which is a coactivator
hypothesized that under specific conditions, such as when of PANX1.67 In periodontal ligament cells, ATP induced the
osteocytes cannot physically touch OC precursors, osteocytes upregulation of RANKL expression via a P2Y1 receptor-
might support osteoclastogenesis by secreting EVs containing cyclooxygenase-dependent pathway.68 Since the release of ATP
RANKL.58 Distinguish the exact localization and form of RANKL from apoptotic cells functions as a specific “find me” message for
produced by osteocytes in vivo is particularly complicated, as the phagocytic cells,69 ATP release during osteocyte apoptosis might
antibodies used for immunohistochemical analyses do not be a specific “find me” signal directed to neighboring osteocytes.
distinguish between the soluble and membrane forms. Therefore, Indeed, ATP from apoptotic osteocytes might bind P2Y2 receptors
to evaluate the form of RANKL that induces osteoclastogenesis, a expressed on bystander osteocytes, which in turn might activate
mouse model was generated with a sheddase-resistant form of RANKL production and release67 (Fig. 2). Moreover, bystander
RANKL with no detectable sRANKL levels in the circulation. osteocytes (in 1–2-mm proximity to the focal damage site)
During growth, this lack of sRANKL did not cause bone mass expressed the antiapoptotic protein BcL-2. This defense mechan-
alteration, but in the adult model mice, the number of osteoclasts ism might be necessary to prevent osteoclastogenic signal
was reduced with an increase in cancellous bone mass.59 damaging viable osteocytes70 (Fig. 2).
However, mice lacking sRANKL expression and ovariectomized As osteocytes are deeply embedded in the bone matrix,
still displayed bone loss because of the lack of estrogen, indicating osteocyte apoptosis or autophagic death causes secondary
that, although mRANKL can support most RANKL functions, the necrosis due to inhibited phagocytosis by scavenger cells.

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
5
Table 1. Systemic and local factors influencing osteocyte function

Stimuli Features affected Factors released Final outcome


(Systemic/Local) (Local) (Local/Systemic) (Systemic/Local)

PTH Life-Span Sclerostin, DKK1, FGF23, DMP1, Phex, MEPE, • Bone Remodeling Balance
Estrogens Network formation Sema 3A RANKL, OPG SASPs, TGF-β1 OCN • Matrix Mineralization
1,25(OH)2D3 Secretome • Perilacunocanalicular Remodeling
Irisin Mechanotrasduction • Errors correction in Ca2+/P homeostasis
Calcitonin • Energy metabolism
Senescence • Myogenesis
TGF-β1 • Oxidative stress
Pi
pO2
Major systemic and local stimuli that influence osteocyte activity, the functions affected, the factors released upon exposure to the stimuli, and the impact on
the system

Necrotic osteocytes release damage-associated molecular pattern osteocytes accompanied by compensatory mineral deposition
(DAMP) proteins after the cell membrane is disrupted.61 DAMP has been observed.82 The same outcome was observed in cases
proteins flow through the canalicular network and reach the of excess PTH concentrations both in rats83 and humans84 and
bone/marrow interface, where they bind to pattern recognition under PTH modulation after exercise.85 This process is mediated
receptors (PRRs) on bone marrow cells.71 When activated by by osteocytes and is called perilacunar remodeling (i.e.,
DAMP–PRR binding, monocytes and macrophages produce “osteocytic osteolysis”); it was initially recognized during lacta-
proinflammatory cytokines, such as TNFα, IL-6, and IL-1.72 These tion. Perilacunar remodeling has been associated with the
cytokines in turn stimulate the expression of RANKL in osteo- activation of PTH-related peptide (PTHrP)–PTHR1 signaling, which
blasts,73 enhancing the osteoclastogenesis previously activated by induces matrix resorptive activity in osteocytes similar to that in
the release of ATP by apoptotic osteocytes.66 DAMPs can directly osteoclasts.86 Perilacunar remodeling integrated with bone
regulate osteoclast formation by activating the membrane-bound remodeling contributes to the maintenance of bone quality.
C-type lectin receptor Mincle. Mincle activation triggers calcium Perilacunar remodeling is an additional component of the long-
signaling and oxidative phosphorylation in osteoclasts, inducing term error correction mechanism to maintain plasma calcium
osteoclast differentiation.74 levels, in addition to the homeostatic regulatory mechanism in
Under physiological conditions, osteocytes exert an inhibitory osteoclasts (RANKL-mediated). The efficiency of this additional
effect on osteoclasts by contributing to the osteoblast production and independent component of bone remodeling in calcium
of OPG via Wnt signaling.75,38 OPG functions as a soluble decoy homeostasis under PTH regulation remains unclear because of
receptor that binds RANKL, preventing its interaction with RANK the lack of a relationship between serum PTH and osteocyte
expressed by osteoclast progenitors. The major cell source of OPG lacuna characteristics.87
is still debated, as it depends on the bone compartment and on Nevertheless, as has been recently discussed,88 osteocytes per
the age of the subject considered. In addition to osteocytes, other sé function independently of associated endocrine loops and
cells can be sources of OPG or RANKL; these cells are exhibit an evolutionary advantage in mineral homeostasis, which
osteoprogenitors, osteoblasts49 or cells of the immune system is particularly highlighted in the rapid minute-to-minute regula-
(B and T cells),76 which are all simultaneously present in a BMU. tion of the BEF-[Ca2+] in teleosts,89 as well as in mammals.26,90
Nevertheless, OPG released by mature osteoblasts and/or Osteocytes, through PTHR1 activation, might counteract age-
osteocytes may constitute the termination signal of the resorp- related bone mass loss. In fact, during aging, as well as under the
tion phase in the remodeling cycle.77 action of several stressors [see Farr et al.91 for a review], senescent
In addition to microcrack-induced signals to activate the healing osteocytes accumulate in the bone microenvironment and acquire
of microdamage foci through targeted remodeling, osteocytes a distinctive proinflammatory secretome, termed the senescence-
sense systemic and local stimuli that affect their functions and associated secretory phenotype (SASP),92 which leads to imbal-
survival. When exposed to a stimulus, osteocytes release several anced bone remodeling with increased resorption and decreased
factors affecting (1) bone remodeling at stochastic loci, (2) matrix formation. When PTHR1 was deleted in mouse osteocytes in vivo
mineralization, (3) lacunocanalicular remodeling, (4) mineral (Dmp1-PPRKO), the affected mice displayed age-dependent
homeostasis, and (5) fat metabolism and myogenesis (Table 1). osteopenia related to a decrease in osteoblast activity with a
parallel rise in osteoclast number and activity. The imbalanced
PTH/PTH-related peptide (PTHrP) bone remodeling in these animals was partially due to a
Under physiological conditions, osteocytes integrate the sclerostin-dependent decrease in osteoblast activity and the lack
responses of bone to mechanical loading and PTH, since the of osteocyte protection from oxidative stress.93
mechanotransduction process requires PTH receptor 1 (PTHR1)
activation to downregulate SOST expression. SOST expression Estrogen
downregulation positively fine-tunes the bone remodeling bal- Osteocytes respond to estrogen by producing the protein
ance by promoting osteoblast expansion.78 When PTHR1 on semaphorin 3A (Sema3A). Sema3A released into the bone
osteocytes is activated by chronically high PTH concentrations or microenvironment binds to its receptor on osteocytes and
when PTHR1 is constitutively active, osteocytes initiate bone promotes their survival. The autocrine loop initiated via Sema3A
remodeling by expanding the osteoclast pool through the release is mainly triggered in the mature cell stage, which is the dominant
of RANKL,79 particularly within the bone cortex.80 In contrast, characteristic of cells in the adult skeleton and is critical in bone
when PTHR1 is deleted, RANKL release and subsequent osteo- remodeling balance, counteracting age-related bone loss. Auto-
clastogenesis do not occur.81 crine loop impairment caused by estrogen deficiency induces an
Interestingly, in addition to BMU activation, three hours after osteoporosis phenotype.94 In addition to its effects on bone
PTH exposure, demineralization of the bone matrix surrounding remodeling, estrogen regulates mechanotransduction in osteocytes

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
6
a
Bone marrow envelope

Pericyte
Bone lining cells
Can
opy

Osteoblast
Monocyte / Macrophage precursor
Microcrack
Primary Remodeling

ts
Osteomorph

as
osteoclast compartment

bl
eo
st
O
Secondary
Re osteoclast
so

id
rb

eo
ed

st
EC

O
M
Reversal cells, RvCs

b Bone marrow envelope

Pericyte Can
opy
Bone lining cells

1. Glucocorticoids
Monocyte/Macrophage 2. Myeloma
Microcrack 3. PostM osteoporosis
Primary 4. Alendronate
osteoclast Osteomorph 5. Aging

Secondary
osteoclast No transition to osteoblasts
Re
so
rb
ed

Reversal phase arrest


EC

Reversal cells
M

Fig. 3 The bone remodeling compartment (BRC). a The BRC provides the correct microenvironment to link bone formation and resorption
through local signaling. The bone marrow envelope (BME) is a layer of cells of mesenchymal origin and a reservoir for osteoprogenitors that
covers the layer of bone lining cells (BLCs). Once remodeling is initiated, osteoclasts lift the BME from the BLC, inducing BME cells to form a
structure called the canopy. The canopy separates the remodeling site from the remainder of the bone marrow to allow osteoclast and
osteoblast precursors to enter the blood compartment. After resorption, osteoclasts either undergo apoptosis or dedifferentiate into
osteomorphs. The resorbed surface is then colonized by secondary osteoclasts and reversal cells. Reversal cells are osteoblast progenitors that
digest fibrillar collagen remnants, similar to BLCs. Secondary osteoclasts and reversal cells provide the basis for the recruitment and expansion
of osteoblastic pools (b) except under certain counteracting conditions, such as glucocorticoid or alendronate treatment, myeloma, or
postmenopausal osteoporosis

by affecting FFSS,95 Wnt/β-catenin expression,96 and Cx43 expres- 1,25(OH)2D3


sion.97 Its removal triggers osteocyte apoptosis and alters the In addition to its systemic effects, 1,25(OH)2D3 binds to VDR in
oxidative microenvironment, leading to the loss of osteocyte osteocytes in an autocrine manner, and as they mature,
resistance to oxidative stress. osteocytes acquire the capacity to convert physiological levels of
25(OH)D to 1,25(OH)2D3.99 The exact role of 1,25(OH)2D3 in
TGF-β1 osteocyte metabolism is still uncharacterized. However, recent
Recently, osteocyte-intrinsic TGF-β1 signaling was discovered as a observations have pointed out that 1,25(OH)2D3 a) stimulates the
regulator of lacuno-canalicular remodeling. The suppression of production of fibroblast factor 23 (FGF23) in osteocyte-like cells;100
TGF-β1 signaling in osteocytes, either pharmacologically (by TGF-β the hormone FGF23 is primarily involved in phosphate home-
receptor type I kinase inhibitors) or genetically (by specific deletion ostasis and vitamin D synthesis; b) modulates matrix mineraliza-
of the receptor TβRII in DMP1-cre mice), leads to the reduction tion by downregulating dentin matrix protein-1 (DMP-1);101 and c)
osteocyte dendrites length and total lacuno-canalicular area. The regulates osteocyte perilacunar remodeling and canalicular
deterioration of the lacuno-canalicular network was accompanied organization through the activation of matrix resorption genes.102
by the decrease in the gene expression of Sost, and metallopro-
teases Mmp2, 13, 14, which are involved in the lacuno-canalicular Phosphate
remodeling, and by the reduction of fracture resistance in cortical The establishment of X-linked hypophosphatemia (XLH) mouse
bone despite no differences in cortical thickness and geometry, models, characterized by elevated serum FGF23 levels, which
and increase in trabecular bone mass, likely due to the observed caused decreased 1,25(OH)2D3 levels and hypophosphatemia, led
decrease in osteoclasts number and surface.98 The observation that to the discovery of the role played by phosphate in osteocyte
lacuno-canalicular remodeling is an essential component of bone functioning, which is triggered in response to disruption of plasma
mechanical competence sustains the view that osteocytes play an phosphate homeostasis.103 How osteocytes sense phosphate
evolutionarily conserved role in bone quality control.98 levels and subsequently regulate perilacunar remodeling103 and

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
7
FGF23 and 1,25(OH)2D3 synthesis104 is still unclear. However, as The generation of the canopy allows the formation of the bone
has been previously discussed,105 the phosphate-sensing mechan- remodeling compartment (BRC), which is considered an essential
ism in osteocytes may involve the activation of FGFR1 and high- component of the BMU itself, as it provides a microenvironment
affinity Na+-Pi cotransporters. to link bone formation and resorption through local signaling.122
The BRC is generated in the proximity of the microvascula-
Calcitonin ture123 (Fig. 3). Although it is unclear how the BRC is formed,
Osteocytes express specific calcitonin receptors, which are several lines of evidence have suggested that the BRC favors a
progressively lost with age.106 Calcitonin can potentially modify direct connection between bone cells and sinusoids, which might
the osteocyte production of sclerostin and FGF23.107 provide reservoirs of osteoprogenitors.122
In support of this hypothesis, an increased presence of capillaries
Irisin covering human cancellous bone has been found at remodeling sites
Irisin, a myokine produced by muscles, reduces osteocyte in an area within 50 µm from the canopy surface.124 This coverage
apoptosis and increases osteocyte number.108 These effects has also been observed on cortical bone remodeling sites.4
suggest a regulatory loop involving osteocytes and muscle The tight link between BRCs and capillaries, which are coated
metabolism, particularly since osteocytes inhibit skeletal muscle with pericytes, allows capillary-BRC canopy interactions to induce
differentiation by producing a large number of cytokines, which BMU development at bone remodeling sites117 (Fig. 3). Pericytes,
might permeate the periosteum or diffuse into the circulation to first identified as cells covering microvessels/capillaries and
negatively regulate myogenesis109. maintaining local homeostasis and angiogenesis, support the
These findings indicate that osteocytes can be considered the mesenchymal niche and can be considered osteoprogenitors.125 It
“social” coordinators of bone cells because they are responsible has been hypothesized that pericytes first move from capillaries to
for maintaining bone physiological responsiveness to mechanical the canopy and then to the bone surface, where they differentiate
and metabolic demands. When osteocytic coordination fails, into mature osteoblasts during bone remodeling.126 It is also
severe osteoporosis develops.110 plausible that the vascularization of the BRC favors the recruit-
ment of osteoprogenitors circulating in the peripheral blood.127
Recently, a specific capillary subtype that supports perivascular
BONE LINING CELLS (BLCS), CANOPY CELLS AND PERICYTES: osteoprogenitor differentiation by producing Nogging via Notch/
COMPOSITION OF THE VASCULAR BONE REMODELING Dll 4 signaling has been termed type H vessel, which express both
COMPARTMENT (BRC) CD31 and Endomucin at high levels. Type H vessels are
BLCs, derived from the final differentiation of osteoblasts, are surrounded by abundant osteoprogenitors (Osterix-positive cells)
located on quiescent trabecular, endosteal, and endocortical and are located both in the metaphysis, close to the growth plate,
surfaces at the end of bone formation. BLCs are a cell population and in the diaphysis (periosteum and endosteum).128,129 During
that differs from both marrow cells111 and osteoblasts. BLCs are bone remodeling, the release of platelet-derived growth factor
flattened and exibits lower synthetic activity, with little cytoplasm type BB (PDGF-BB) from preosteoclasts induces an increase in
or endoplasmic reticulum,112 but they retain a social attitude. endothelial cell recruitment and assembly, with a subsequent rise
Similar to osteocytes, BLCs express intercellular adhesion in the number of type H vessels. Simultaneously, PDGF-BB triggers
molecule-1 (ICAM-1) to maintain functional contact with osteo- the recruitment of osteoprogenitors from vessels to the bone
blasts, osteoclast precursors113 and mature osteoclasts.114 They surface.130 The formation of H vessels is supported by the
cover quiescent bone surfaces that are not undergoing remodel- production of the slit homolog 3 (SLIT3) protein in osteoclasts,131
ing, but they are not “quiescent” as the older literature suggests. which also enhances H vessel branching.131–133
Although their specific function has not yet been fully defined, Type H vessels respond to the administration of intermittent
BLCs might form an epithelial-like membrane that functions as an anabolic PTH (iPTH), which promotes the detachment of leptin
ion partition system between bone and systemic extracellular receptor-expressing (LepR+) cells, which are pericytes that can
fluids in Ca2+ homeostasis.26,90,115 Indeed, the BLC membrane differentiate into osteoprogenitors,134 from CD31hi/Edmhi vessels.
expresses tight junction membrane proteins that are responsive to Pericyte migration contributes to the osteoprogenitor recruitment
metabolic demands such as chronic metabolic acidosis.116 More- to the BRC during PTH-induced bone remodeling.135 Notably,
over, BLCs stain positive for osteoblast markers such as alkaline anabolic responses to iPTH as well as sclerostin inhibitor
phosphatase, (ALP) osteocalcin (OCN), and osteonectin, in treatments, decline with time,136 as it is conceivable that the
agreement with their osteogenic potential. BLCs are covered by precursor pool tends to be exhausted after major stimulation.
a thin layer of mesenchyme-derived cells called bone marrow BLCs do not exclusively represent a final differentiation stage of
envelope (BME) cells that morphologically resemble BLCs and, the osteogenic line; they can also be osteoblast precursors.137 A
similar to BLCs, are considered osteoprogenitors.2 study by Matic et al.137 led to the development of a model in
When the remodeling starts, BME cells and BLCs form a which osteoclasts initiate osteoprogenitor cell expansion (see
protective structure known as the canopy, which separates following section) by activating osteoprogenitor reservoirs located
osteoclasts and osteoblasts from bone marrow117 (Fig. 3). At the in proximity to the eroded bone surface consisting of BLCs,
bone remodeling site, BLCs disconnect from the underlying canopy cells and pericytes.120
osteocytes through gap junction disruption, and digest fibrillary Hence, BLCs favor the initiation of bone formation by supporting
collagen, the most abundant component in the bone extracellular the recruitment of bone-forming osteoblasts from the canopy
matrix (ECM), which is tightly packed in a helical structure to upon the release of osteoclastic factors supporting bone formation,
provide mechanical stability and confer resistance against independently of bone resorption (see following section). The
proteolysis.118 To be degraded, fibrillary collagen usually requires osteoclast-canopy interface is therefore the physical site where the
the activation of the cysteine protease cathepsin K and members coupling of bone resorption to bone formation occurs.117
of the matrix metalloproteinase (MMP) family.119 While osteoclasts
express both of these enzymes types, which contribute differently
to bone resorption,120 BLCs do not express cathepsin K but can OSTEOCLASTS
efficiently remove nonmineralized fibrillary collagen (present Resorbing osteoclasts: osteoclastogenesis and excavation of
either on the quiescent bone surface or in resorption lacunae resorption lacunae
after osteoclastic activity)113 mediated through their highly active Osteoclasts are responsible for bone resorption, a process that is
MMPs.121 accomplished in a fairly short time relative to that required for

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
8
bone formation. Osteoclasts originate from the hematopoietic sRANKL or mRANKL produced by osteogenic cells binds to RANK
monocyte-macrophage lineage, residing within the granulocyte- on osteoclast precursors to activate intracellular signaling of tumor
macrophage colony-forming unit. Osteoclastogenesis starts in the necrosis factor receptor-associated factors (TRAFs), especially TRAF
bone marrow upon the release of RANKL and macrophage colony- 2, 5, 6, which are adapter molecules that trigger NF-κB
stimulating factor (M-CSF) from osteocytes and vascular endothe- activation.162 NF-κBs are transcription factors that were originally
lial cells close to the bone surface.138 It then extends toward the discovered as regulators of B lymphocyte differentiation163 and
bone surface through the layer of BLCs along an “osteogenesis that play important roles in innate and adaptative immune
route”, likely guided by physical (i.e., collagen) and biochemical responses.164 Mice lacking both NF-κB 1 and 2 display, among
(i.e., sphingosine-1-phosphate, S1P) signals.139 In vitro experi- other features, severe osteopetrosis165 due to an accumulation of
ments have shown that osteoclasts access bone surfaces by osteoclast precursors.166 NF-κBs can activate canonical signaling
directly displacing the cells present on the bone surface.140 TGF- and noncanonical signaling pathways in osteoclasts. Canonical NF-
β1 is important for inducing cytoskeletal reorganization via the κB signaling activation is very fast, initiated within one hour of
p38 pathway in osteoblastic cells, which elongate to generate cell- RANKL binding, and leads to the induction of NFATc1 expres-
free areas.141 The inhibition of c-src and subsequent actin sion.167 NFATc1 is a transcription factor that plays a master role in
organization in osteoclast precursors prevent osteoclasts from osteoclastogenesis168 by regulating the expression of osteoclast-
migrating through the osteoblast layer.142 Metalloproteinase specific genes such as cathepsin K, DC-STAMP, and tartrate-
(MMP) inhibitors prevent both osteoblast elongation and the resistant acid phosphatase (TRAP) or that of osteoclast-associated
subsequent generation of cell-free areas143 and directly act on receptor (OSCAR) and other genes involved in OC resorptive
osteoclasts by preventing their migration through the cell layer.141 functions.169 The role of the Nf-κB noncanonical pathway in
However, in osteoclasts, MMP inhibitors did not affect actin osteoclastogenesis is controversial. Global deletion of single
organization per sé,142 although they had been previously molecules involved in the Nf-κB noncanonical pathway did not
demonstrated to prolong the podosome lifespan in these cells.144 affect osteoclast numbers in vivo,170 whereas conditional deletion
MMP inhibitor administration fully prevented osteoclast recruit- of TRAF3, an inhibitor of the noncanonical pathway, in osteoclast
ment in the diaphysis core in vivo145 as well as osteoclast precursors resulted in increased osteoclast formation and bone
migration through collagen in vitro.146 Among MMPs, MMP-14 resorption in mice.171
and MMP-9 seem to play a major role in osteoclast migration. The discovery of a forward signaling induced by RANKL binding
MMP-14, also known as MT1-MMP, is expressed in all skeletal cells to its receptor RANK on osteoclast precursors led to the
but is more abundant in osteoclasts.147 MT1-MMP-knockout (KO) development of denosumab (DMAB), a human monoclonal
mice usually die couple of months after birth, and display a severe antibody that binds RANKL and inhibits osteoclastogenesis.
phenotype that includes delayed ossification, unclosed sutures Recently, a second receptor for RANKL, the leucine-rich repeat-
and unremoved cartilage.148 MMP-14 is highly expressed on containing G protein-coupled receptor LGR4, was discovered on
osteoclast podosomes,149 and it digests interstitial collagen as well the osteoclast membrane. The extracellular domain of LGR4 binds
as other ECM molecules.150 MMP-9-KO mice showed reduced RANKL and inhibits NFATC1 expression, thus blocking osteoclas-
osteoclast invasion into the core of developing bones,151 and togenesis.172 Very recently, a variant of RANKL with changes in a
in vitro, osteoclasts lacking MMP9 did not migrate.152 However, few amino acids in the RANK binding site was developed. This
very recently, it has been demonstrated that osteoclasts lacking variant still activates LGR4 signaling and inhibits osteoclastogen-
either MMP9 or MMP14 alone (in this case conditionally deleted in esis. Moreover, it has been observed that this variant acts as an
the myeloid population) did not show altered fusion and function, immunogen triggering the production of anti‐RANKL antibodies,
or any defects in bone resorption. Only the deletion of both MMP9 and its use might reduce the risks linked to the abrupt suspension
and MMP14 generated a reduction in bone resorption areas and in of denosumab use.173,70 The balance achieved among RANK,
type I collagenolysis activity.153 RANKL, OPG, and LGR4 fine tunes bone resorption in the bone
The fusion of the earliest osteoclast precursors into multi- remodeling process, and the pharmacological modulation of this
nucleated, mature osteoclasts is a critical process in osteoclasto- balance has critical therapeutic potential in osteoporosis.172
genesis, as the number of nuclei per cell determines osteoclast In addition to RANKL/RANK, LGR4 and M-CSF signaling174
aggressiveness.154 The presence of distinct giant hypernucleated guiding and balancing the early steps of osteoclastogenesis, other
osteoclasts per sé in patients undergoing long-term treatment factors act in concert to determine the osteoclastic mature cell
with nitrogen bisphosphonates (NBPs), as discovered by Weinstein phenotype, including β3-integrin,175 NR4A1,176 immunoreceptor
et al.155 and confirmed by others,156 did not necessarily indicate tyrosine-based activation motif-containing proteins (ITAMs),
an increase in osteoclast aggressiveness. These osteoclasts indeed DNAX-activating protein and the Fcγ receptor.177 Additionally,
showed decreased resorption competency and prolonged apop- Toll-like receptors (TLRs) expressed on osteoclast progenitors178
tosis characteristics. A recent study with transmission electron promote differentiation toward the mature cell phenotype, as
microscopy demonstrated that NBPs first induce osteoclast observed in inflammatory osteolytic diseases.179
apoptosis and then the formation of newly differentiated WNT proteins such as Wnt5A in the noncanonical pathway have
osteoclasts that can anchor the bone surface but cannot form a been outlined as key elements in osteoclastogenesis. WNT5a
clear zone or ruffled border.157 NPBs induce osteoclasts to acquire produced by osteoblasts supports osteoclastogenesis by stimulat-
a nonresorbing phenotype by inhibiting c-Src, which is essential ing RANK expression in osteoclast precursors,180 promoting
for cytoskeletal construction,158 and by stimulating dendritic cell- osteoclast fusion,181 and supporting actin ring formation and
specific transmembrane protein (DC-STAMP),159 which is neces- bone resorption via c-Src.182 Since WNT5a enhances osteogenic
sary for cell fusion. pathway of Wnt/β-catenin activation in osteoblasts,183 it was
Under physiological conditions, osteoclast fusion requires thought that WNT5a produced by osteoblasts might affect both
immobility and heterogeneity between fusion partners; fusion resorption and formation. However, recent studies have led to
can only occur at the bone surface and must be completed for modifications of this previous assumption as they demonstrated
successful resorption.139 As reviewed elsewhere,160 heterogeneity that, after RANKL stimulation, osteoclast precursors also produce
is an integral feature of osteoclast fusion and may be related to and secrete a unique phosphorylated form of WNT5A. Further-
the capacity of fusion precursors to adapt to specific bone more, deletion of this phosphorylated form of WNT5a in mature
microenvironments. Fusion also requires RANKL, which is highly osteoclasts led to reduced bone formation in male mice.184 Hence,
expressed by BLCs;161 osteoblasts in the growing skeleton; and it is speculated that in the BRC, osteoblast WNT5a first supports
osteocyte arborizations that reach the bone surface in adults.51,52 osteoclastogenesis; then, as the osteoclast precursors differentiate,

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
9
Osteoblast precursors

TGF-β1

IGF-1

MIGRATION
Wnt1
TGF-β1
Wnt1 DIFFERENTIATION BMP2, Wnt10b
RUNX2,
OPN, DMP1,
IGF-1 PHEX, SOST
TGF-β1 receptor

IGF-1 receptor
Osteoblasts
Osteoclast
Wnt receptor

Resorption lacuna
MINERALIZATION
ECM Wnt10b

Fig. 4 Released extracellular matrix (ECM) factors. Two major factors are released by the ECM upon bone resorption and cooperate to regulate
bone remodeling and BMU activity: TGF-β1 and IGF-1. After release from the matrix, active TGF-β1 acts both on osteoblasts and osteoclasts to
induce osteoblast precursor migration to the site of resorption and osteoclast production of Wnt1, which promotes osteoblast recruitment
and/or differentiation at sites of bone resorption, and Wint10b, which promotes matrix mineralization. IGF-1 supports the recruitment of
osteoblast progenitors and promotes osteoblast differentiation and matrix mineralization by inducing the transcription of osteogenesis‐
related genes such as DMP1, PHEX, SOST, BMP2, RUNX2, OPN, and OCN

WNT5a expression increases, leading to the subsequent enhance- also providers of multiple coupling signals196 for osteoblast
ment of osteoclast fusion and activity. In later stages, WNT5a formation.
production by mature osteoclasts fosters osteoblastogenesis
because WNT5a cooperates with canonical Wnt signaling. The Resorbing osteoclasts: the release of bone matrix factors
subsequent expansion of the osteoblast pool might therefore The bone ECM resorbed by osteoclasts contains inorganic and
fulfill the resorption cavity at the proper time and space, organic compounds. A growing body of evidence in ECM biology
suggesting that Wnt5a is a clastokine that maintains suggests that several organic proteins of the matrix regulate the
osteoclast–osteoblast coupling in physiological bone remodeling. quantum of BMU activity. While the only inorganic components
The cytoskeletal and membrane organization of osteoclasts of the ECM are apatite and trace elements, proteomic analysis
characterizes their differentiation state. Mature osteoclasts are have led to the identification of more than 100 organic ECM
polarized cells with an apical membrane facing bone and an proteins,197 with collagen type I (Coll-1) and noncollagenous
opposite basolateral membrane facing plasma. Both apical and proteins (NCPs) being the major constituents. NCPs can bind
basolateral membrane domains are essential structures for growth factors, membrane receptors, and adhesion molecules,
resorbing osteoclasts.185 Critical functional features of the apical forming an intrinsic biochemical signaling network within each
membrane domain are the sealing zone and the ruffled border. BMU. NCPs comprise several Gla proteins, including bone-Gla
The sealing zone establishes contact with the bone surface and protein (i.e., Osteocalcin, OCN), matrix-Gla protein (MGP),
delimits the resorption lacunae (i.e., Howship’s lacunae). The protein-S, Gla-rich protein (GRP), periostin, and periostin-like
ruffled border membrane allows large vesicle transport, the factor (PLF), as reviewed elsewhere.198 In addition to OCN, whose
delivery of hydrochloric acid (by vacuolar-type H + ATPase186) hormonal effects were previously established in animal mod-
and chloride ion channel-7 (CLC-7),187 and the release of several els,199 Gla proteins exhibit several functions supporting bone
lysosomal proteases, such as cathepsin K, TRAP, and MMPs.188 metabolism through γ-carboxylation-dependent and γ-carbox-
After endocytosis, the products derived from matrix degradation ylation-independent mechanisms, ranging from the regulation
are transported through the cell within vesicles for further of cell adhesion and activity to the modulation of calcium
intracellular degradation and exocytosed on the opposite site concentrations in the extracellular space. Gla proteins act in
through the secretory domain at the basolateral membrane.189 concert with glycoproteins such as osteonectin, thrombospon-
Thus, the osteoclasts never lose their tight attachment to the bone dins and R-spondins, sialoprotein and matrix extracellular
surface when resorption occurs. The interaction with the matrix is phosphoglycoprotein (MEPE) and DMP1 to control mineraliza-
mediated by the integrin complex α5β3, which recognizes RGD tion, synergistically with FGF23 and PHEX produced by
motifs in proteins such as fibronectin,190 osteopontin (OPN)191 and osteocytes (see Lin et al.200 for review).
bone sialoprotein.192 By binding to RGD motifs, the α5β3 complex Two major factors released by the ECM influence bone
is activated and clusters at the ruffled border,193 thus allowing the remodeling and BMU activity: transforming growth factor-β
activation of focal adhesion complexes and the formation of the (TGF-β) and insulin growth factor-1 (IGF-1). Together, TGF-β and
sealing zone during resorption.194 It has been hypothesized that IGF-1 influence the recruitment and differentiation of osteoblast
integrin β3 contains two motifs with different affinities (high and lineage cells (Fig. 4), favor bone matrix mineralization, and
low) for Ca2+ concentrations. A normal Ca2+ concentration in BEF regulate osteoclast activity. For these reasons, they are acknowl-
maintains low-affinity domain activation, which does not maintain edged as “coupling factors”.
a strong attachment between β3 and RGD. When Ca2+
concentrations diminish, such as when osteocytes are injured, TGF-β. TGF-β1 is one of the most abundant cytokines in the
the high affinity site is the only one activated; this motif induces bone matrix.201 TGF-β1, which is a member of the TGF superfamily
αvβ3 clustering, which allows osteoclasts to strongly adhere to the that includes bone morphogenetic proteins (BMPs), binds to
matrix and initiate bone resorption.195 specific TGF receptors (TGFR1-2) in osteoblasts, where it activates
The overall fate of osteoclasts (i.e., recruitment, differentiation, the synthesis of collagen and the expression of RUNX2, a master
fusion and apoptosis, see below), is tightly controlled by the transcription factor regulating osteoblast proliferation and differ-
process coordinating bone remodeling, in which osteoclasts are entiation. TGF-β1 release and activation, resulting from the activity

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
10
of metalloproteinases and interaction with integrins, are pre- action might be limited to the early phases of remodeling,
requisites for the exertion of TGF-β1 effects on bone cells. This when osteoclasts are still actively resorbing the bone matrix.
occurs during bone resorption since osteoclasts lower the pH in
resorption lacunae and secrete cathepsin K, thus releasing and
activating TGF-β1.202 When osteoclasts actively resorb bone, TGF- ANABOLIC OSTEOCLASTS: A BRIDGE FROM BONE RESORPTION
β1 is released in the lacunae, thus playing a crucial role in the TO FORMATION
balance between bone formation and resorption. First, active TGF- The resorption of the bone ECM is necessary, but not sufficient, to
β1 induces osteoblast precursor migration to the site of induce the stimulus provided by osteoclasts to osteoblasts during
resorption203 without affecting the differentiation of cells.204 Since remodeling. Currently, available evidence indicates that osteo-
TGF-β1 alone is not able to promote osteoblast differentiation,205 clasts can support bone formation independent of their resorption
cooperative pathways need to be activated to foster an activity. The paradoxical concept of “anabolic osteoclasts” was
osteogenic environment at resorption surfaces within the BRC. A articulated after the original observation that human osteoblasts
cooperation pathway is provided by the osteoclast itself. In exposed to conditioned media from human osteoclasts increased
addition to activating and releasing TGF-β1 from the bone matrix, bone nodule formation.221 The finding that the transplantation of
osteoclasts express specific receptors for TGF-β1. Osteoclastic TGF- nonfunctional osteoclasts in irradiated skeletally mature 3-month-
β1 receptor signaling stimulates the production of Wnt1, which old wild-type mice induced a high trabecular bone volume,
promotes osteoblast recruitment and/or differentiation at sites of increased bone strength, and an increased bone formation rate in
bone resorption. Since impaired TGF-β1 receptor signaling in trabecular bone222 supports this concept. The anabolic role of
osteoclasts has detrimental effects on bone mass in mouse osteoclasts can be observed in both humans and mice with
models, it is likely that the release of Wnt1 constitutes a coupling osteoclast-rich osteopetrosis (OPT), a disease resulting from
pathway.206 Moreover, TGF-β1 favors RANKL production in mutations of either the V-ATPase subunit or the ClC-7 chloride
osteoblast precursors by inducing TRAF3 degradation. The channel that abrogate the osteoclastic acidification process, which
expression of TRAF3, which is an inhibitor of noncanonical Nf-κB is essential for resorption activity. Under these conditions, no
signaling, in osteoblast precursors favors the stabilization of factors stored in bone were released from the unresorbed bone
β-catenin and, therefore, their differentiation into mature osteo- matrix, but the long-surviving osteoclasts, fused into large
blasts. In the absence of TRAF3 expression, Nf-κB noncanonical abnormal multinucleated cells, provided an osteogenic milieu.223
signaling is activated with an increase in RANKL production and In fact, in models of osteoclast-rich OPT, bone formation is
therefore is associated with osteoclastogenesis.207 With matrix maintained or enhanced despite the impairment of resorption
resorption progression, the concentration of TGF-β1 increases, and activity,224 thus providing evidence that nonresorbing osteoclasts
TGF-β1 activates a negative feedback loop in osteoclastogenesis. promote/maintain bone formation. Findings following the phar-
High TGF-β1 expression inhibits the migration of osteoclast macologic blockage of cathepsin K in osteoclasts (Odanacatib) are
precursors208 by suppressing RANKL expression and by stimulat- in line with this view. In patients treated with Odanacatib,
ing OPG production in osteoblasts.209 A reduced RANKL/OPG ratio although osteoclast bone resorption was impaired, the osteoclast
indeed results in reduced osteoclastogenesis. In the absence of number was increased to be more than twofold higher than that
osteoclast survival factors such as RANKL, TGF-β1 leads to after placebo treatment by month 60, and bone formation, after a
osteoclast apoptosis via the upregulation of Bim expression.210 transient reduction, was maintained.225
This proapoptotic effect of TGF-β1 on osteoclasts is consistent In contrast to “osteoclast-rich” OPT, active bone formation is
with the crucial role it plays in the reversal of bone resorption to lacking in “osteoclast-poor” OPT (due to TNFRSF11A mutations226)
bone formation. Indeed, TGF-β1 release enhances Wnt10b or dysosteosclerosis (DSS) (an OPT of unknown etiology227).
expression and secretion to stimulate osteoblast-directed miner- Indeed, bone histomorphometric analysis showed no linear single
alization. TGF-β1 therefore has a dual effect on osteoblasts by or double tetracycline labels under these conditions.
directly recruiting their progenitors to the bone surface and by Therefore, these “anabolic osteoclasts” might produce a pool of
indirectly promoting matrix mineralization through osteoclast- signals that activate bone formation and that can be a) released
derived Wnt10b.211 into the BRC, b) shuttled to osteoblasts, and c) transferred by
receptor binding.
IGF1. IGF-1 is the most abundant growth factor deposited in
the bone ECM.212 Osteocytes in particular secrete large Information released into the BRC
amounts of IGF1, which is incorporated in the bone matrix.213 Several osteoclast-derived factors released into the BRC have been
Since IGF-1 is produced under loading, it might contribute to identified and validated in genetically modified mouse models.
the mechanotransduction process.214 Whether released by cell These factors include cardiotrophin-1 (CT-1),228 S1P,229 Wnt10B,
secretion or bone ECM degradation, IGF1 is a local autocrine/ BMP6,230 collagen triple helix repeat-containing 1 (CTHRC1),231
paracrine regulator of bone remodeling and does not con- complement factor 3a (C3a),232 and leukemia inhibitory factor
tribute to the circulating IGF-I pool. IGF-1 acts primarily as a (LIF).233 However, not all of these identified factors are responsible
migratory signal and as a differentiation factor for osteoblast for coupling in bone remodeling, nor are they produced only by
precursors, rather than as a proliferation factor. IGF-1 recruits osteoclasts due to macrophage contamination in vitro and the
progenitor cells of the osteoblastic lineage215 and promotes presence in vivo of immunocompetent cells within the remodel-
osteoblast differentiation and matrix mineralization by upregu- ing compartment. Whether these factors exhibit similar effects in
lating the osteogenesis‐related genes DMP1, PHEX, SOST, BMP2, humans remains unclear; however, a recent study shed some light
RUNX2, OPN, and OCN.216 Moreover, IGF-1 released from the on this controversy. Bone biopsy samples taken from postmeno-
bone matrix during bone resorption establishes an osteogenic pausal women treated with DMAB were analyzed by RNA
microenvironment by activating mechanistic target of rapamy- sequencing, and the results were compared to those obtained
cin (mTOR) signaling,217 which is critical for cellular energy from bone biopsy samples of untreated subjects. The comparison
metabolism and cell migration.218 Recent studies have con- revealed potential osteoclast-derived coupling factors in humans:
firmed that IGF-1-dependent activation of mTOR induces LIF, CREG2, CST3, CCBE1, and dipeptidyl peptidase-4 (DPP4), a
human dental pulp stem cell (DPSC) differentiation toward an highly conserved cell surface peptidase. The reduction in
osteoblastic phenotype.219 osteoclast-derived DPP4 in the DMAB-treated group was asso-
Since TGF-β1 and IGF1 do not remain accessible long enough ciated with a significant increase in glucagon-like peptide-1
to affect the complete refilling of the resorption cavity,220 their (GLP)−1 in the serum compared to that in the placebo-treated

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
11
that vesicular RANK binds to RANKL on the osteoblast surface (and
possibly on osteocytes) and triggers the mTOR-dependent
Monocyte/Macrophage
stimulation of Runx2238 (Fig. 5). This finding suggests a previously
RANK unknown scenario in which actively resorbing osteoclasts promote
the differentiation of neighboring osteoblast precursors into
RANKL mature bone-forming cells. The physiological importance of the
messages shuttled by EVs has been confirmed by the observation
Forward signaling RANKL that osteoblasts do not efficiently deposit bone ECM when RANKL
OC
reverse signaling is suppressed.238
OB The discovery of the reverse signaling induced by RANK on
Apoptotic OC EphB2 EphB4 R
TO osteoclasts binding to RANKL on osteoblasts indicates new
m
pharmacological possibilities. Interestingly, the administration
of a specific peptide, W9, to prevent RANKL-RANK signaling has
been shown to exert a bone anabolic effect in vivo by inducing
RvCs
uncoupling.239 Therefore, DMAB may activate RANKL reverse
RANKL
vesicles signaling,240 explaining the continuous increase in bone
Reverse signaling
Apoptotic mineral density (BMD) observed at the 10-year follow-up of
bodies DMAB-treated individuals.241 However, experimental evidence
supporting the hypothesis of the bimodal effects of DMAB on
RANKL-RANK signaling during bone remodeling is lacking
because remodeling-based bone formation is severely sup-
pressed in the femur of DMAB-treated subjects.242 It is more
Fig. 5 Anabolic osteoclasts. Osteoblasts stimulate osteoclastogen-
esis by producing RANKL as a forward signal. However, osteoclasts
likely that DMAB blocks RANKL expressed on bone marrow
can act as anabolic cells by generating positive reverse signaling in mesenchymal stem cells (BMSCs),243 through which it nega-
osteoblasts. Both resorbing and apoptotic osteoclasts can release tively regulates osteogenic differentiation.244 According to a
extracellular vesicles (EVs) that contain RANK. Once discharged from hypothesis suggested by Wang et al.,243 the DMAB-related
EVs, RANK binds RANKL clusters on the osteoblast membrane and abrogation of RANKL-induced inhibition of osteoblast precur-
activates osteogenesis via the mTOR pathway. In addition, sors might underlie the activation of modeling-based bone
EphrinB2 signaling from osteoclasts to EphB4 on osteoblasts/bone formation242 and sustain the increase in BMD over time, as
lining cells (BLCs)/reversal cells favors osteogenic differentiation previously observed.241
RANKL monomers expressed on osteoblasts are not activated
group, suggesting that DPP4, in addition to its function as a by RANK238 or by OPG, as indicated by the deletion of RANK or
coupling factor, might constitute a potential link between bone OPG failing to reduce bone formation.245 In contrast, the
remodeling and energy metabolism.234 multimeric assembly of RANKL can activate reverse signaling. In
Mature human osteoclasts also secrete SLIT3, a chemorepellent fact, OP3-4, a RANKL-binding peptide that induces the clustering
originally identified as a regulator of axon crossing in the brain. of RANKL on the cell membrane, induces the activation of RANKL
Osteoclast-derived SLIT3 stimulates the migration and prolifera- reverse signaling.246 RANKL clustering on early osteoblasts may
tion of osteoblast lineage cells via the activation of β-catenin and therefore be required for the activation of RANKL reverse signaling
suppresses osteoclastogenesis in an autocrine manner.131 SLIT3 in resorption lacunae, similar to the effect of shuttling RANK in
contributes to the initial establishment of the osteogenic osteoclast EVs during bone remodeling (Fig. 5) or that of an anti-
environment after resorption and meets the requirements to be RANKL antibody-containing leucine zippers, which induce trimer
considered a coupling factor, as i) SLIT3 production increases formation.238
during osteoclast differentiation and ii) SLIT3 is necessary for Some of the exosomes released by osteoclasts at the end of
osteoblast precursor migration.235 Considering that bone greatly the two-week cell life span contribute to signal transfer during
contributes to SLIT3 levels in plasma, SLIT3 might be considered a remodeling. In fact, apoptotic osteoclasts produce large amounts
novel biomarker of bone turnover and a candidate for the of apoptotic bodies. Apoptotic bodies are subcellular membrane-
treatment of osteoporosis because it plays active roles in both bound EVs containing fragments of nucleus, endoplasmic
bone resorption and formation through its opposite effects. reticulum and mitochondria that assemble in a random manner
and are involved in intercellular crosstalk.247 Osteoclast-derived
Information shuttled to osteoblasts apoptotic bodies contain osteoclast differentiation factors, such
The coupling of osteoclasts to osteoblasts and related functions as RANK and RANKL (Fig. 5). In contrast, osteoblast-derived
involve not only the secretion of factors in the BRC but also the apoptotic bodies contain specific osteogenic factors, such as
production of specific EVs, released by bone cells. EVs have BMPs, OPN, OCN and bone ALP (BALP). By mapping the whole-
emerged as important intercellular regulators since they function proteome signatures of osteoclast-derived apoptotic bodies, the
as messengers from osteoclasts to osteoblasts, and vice versa.236 apoptotic bodies content was confirmed to be consistent with
EVs exhibit an evolutionary advantage over the paracrine that of their parent cells in terms of both proteome signatures
pathway activated by released factors because they can protect and biological functions.248 During remodeling, osteoclast-
their message (mRNAs and proteins, including cytokines, etc.) derived apoptotic bodies containing miR-214-3p are delivered
from degradation and might therefore play critical roles in fine- to osteoblasts and serve as intercellular messengers that inhibit
tuning bone remodeling. osteoblast differentiation and subsequent bone formation.249
Osteogenic cells trigger hematopoietic precursor differentiation Recently, the roles of osteoclast-derived apoptotic bodies have
into osteoclasts by secreting RANKL. In addition, through a less- been more precisely characterized, and their effects have been
characterized mechanism, osteoclasts stimulate osteoblasts using found to be determined by the differentiation state of parental
the same RANKL–RANK pathway acting in reverse mode; that is, cells.250 For example, apoptotic bodies derived from mono-
RANKL acts as the receptor. In fact, osteoclasts release EVs nuclear preosteoclasts undergoing apoptosis deliver PDGF-BB to
(between 25 and 120 nm in diameter) that carry RANK. These recipient endothelial progenitor cells (EPCs), and since PDGF-BB
vesicles were initially thought to be negative paracrine regulators is a proangiogenic factor,130 preosteoclast derived apoptotic
of osteoclastogenesis.237 However, a recent study demonstrated bodies promote angiogenesis. However, apoptotic bodies

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
12
derived from multinucleated mature osteoclasts promote osteo- highly motile osteoclast daughter cells that remain in the adjacent
genesis via RANKL reverse signaling. bone marrow and retain the ability to fuse back into functional
Therefore, in addition to well-studied osteoclast–osteoblast osteoclasts.257
coupling, the involvement of osteoclast-derived apoptotic The discovery of a transient cell stage of osteoclasts (i.e., the
bodies promotes the transition from bone resorption to osteomorph) that can undergo phenotype reversion, analogous to
formation during bone remodeling. Although the mechanism what occurs in osteoblasts (i.e., BLCs), implies the presence of two
by which apoptotic bodies delivery is controlled within each reacting components of the BMU that can rapidly adjust the
BMU is unclear, mature osteoclasts derived apoptotic bodies operational process of bone remodeling. This implication is
further connect the disappearance of osteoclasts from resorption clinically relevant to osteoporosis therapy. For example, BLCs
lacunae with the incoming osteogenic process involving can react to PTH exposure by quickly reverting to osteoblasts and
osteoblast recruitment, migration and differentiation, contribut- thus expand the osteoblastic pool258 to fill resorption lacunae,
ing to the reversal of cellular activity in reverting from bone similar to process in the “anabolic window” after teriparatide and
resorption to bone formation. abaloparatide administration.259 Similarly, exposure to RANKL
might allow osteomorphs to rapidly fuse and recycle back into
Information transferred by receptor binding functional osteoclasts. Since RANKL inhibition by DMAB results in
The communication route between osteoclasts and osteoblasts osteomorph accumulation,257 the reconstitution of RANKL signal-
involves a bidirectional signaling pathway mediated by EphrinB2‐ ing, as occurs upon DMAB discontinuation, can activate massive
EphB4 that links the suppression of osteoclastogenesis to bone resorption over a very short time scale, leading to rebound
osteogenesis within each BMU.251 EphrinBs are transmembrane fractures.260 This rebound effect is not fully inhibited by bispho-
proteins with cytoplasmic domains that are preferential ligands for sphonate therapy,261 probably because osteomorphs first accu-
the tyrosine kinase receptor EphB. EphrinB2 expression in mulate at bone sites and are thus not accessible to potent
osteoclasts is associated with RANKL-induced osteoclast differ- bisphosphonate treatment. After osteoclasts are no longer active
entiation, whereas EphB4 is constitutively expressed in osteo- and disappear, the Howship’s lacunae remain covered with
blasts. Signaling from EphrinB2 in mature osteoclasts to EphB4 in undigested nonmineralized collagen matrix. Mononucleated cells
osteoblast precursors stimulates osteogenic differentiation (Fig. 5), colonize these eroded surfaces (ESs), covering 80% them.262 These
whereas signaling from EphB4 in osteoblasts to EphB2 in cells, called reversal cells,263 lack specific cell identification
osteoclasts inhibits the differentiation of osteoclast precursors. markers. The available literature has identified the cells appearing
EphrinB2–EphB4 bidirectional signaling must function locally and at the resorption lacunae as osteoclasts264 or, more recently, as
requires direct cell‐cell contact since both receptors are anchored osteoblasts.265 Indeed, the reversal cells express osteoblast lineage
to the cell membrane, as shown with goldfish scale model of bone cell markers such as Runx2, ALP, and Col3, but not TRAcP or
remodeling.252 In humans, the role of EphrinB2 and EphB4 in bone CatK.265 Indeed, reversal cells express osteoblast markers such as
remodeling has been debated because osteoclasts actively Runx2, ALP, and Col3 but not TRAcP or CatK.265 Abdelgawad
resorbing bone, and osteoblasts forming bone do not directly et al.266 showed that the immunoreactivity of these cells for
get in contact in the BRC. As a result the cell–cell interaction osteoclast-derived TRAcP is attributable to TRAcP taken up by
mediated by EphrinB2 and EphB4 might be limited to the osteoblast lines from neighboring osteoclasts and does not
precursor stage, when osteoclastogenesis and osteoblastogenesis represent the acquisition of an osteoclastic phenotype. Moreover,
are simultaneously regulated, or (as suggested previously253) to Abdelgawad et al.266 observed that early reversal cells, located in
the mature osteoclast stage, in which direct contact is realized close proximity to resorbing osteoclasts and forming direct cellular
with the BLCs surrounding the canopy. Under PTH1R activation, contacts with neighboring osteoclasts through short cytoplasmic
EphrinB2 expressed on osteoclasts has been suggested to affect extensions, are osteoblast progenitors with the capacity to digest
EphrinB4 on the osteoblast/BLC surface to induce osteoblast fibrillar collagen remnants, similar to BLCs.113 In particular, reversal
commitment and promote osteoblast differentiation.254 cells have been shown to be distributed among osteoclasts in the
resorption surface far from the pockets of osteoclasts at the
cutting cone tip. New resorption events might occur after the
INTERMEDIATE CELL PHENOTYPES: OSTEOMORPHS AND primary excavation of the resorption cavity/canal when reversal
REVERSAL CELLS TUNING AND COUPLING BONE REMODELING cells have colonized ES.256 In contrast, osteoclasts have never
The orderly genesis and apoptosis of both osteoclasts and been observed at the bone formation wall, suggesting that bone
osteoblasts are essential for physiological bone homeostasis resorption continues until bone formation takes place. Lassen
during bone remodeling. Possible intermediate phenotypes may et al.’s256 findings indicate that a pure reversal period, as
be appropriate targets for coupling factors by providing a temporal commonly depicted following a halt in resorption,8 does not take
connection between osteoclasts and osteoblasts. In the traditional place. In contrast, it is conceivable that the expanded pool of
view, osteoclasts were thought to live for two weeks11 before reversal cells, initially interacting with bystander osteoclasts in the
undergoing apoptosis; it was therefore assumed that during bone same time and space, gradually switch off further resorption and
remodeling, the osteoclast resorption phase within a single BMU form an osteogenic environment, activating bone formation later,
was temporally limited by the osteoclast life span, ending with the when osteoclasts are completely absent.
completion of its bone-resorbing activity. However, the observa- The dynamic events described here support the view that the
tion that osteoclasts can survive longer by fusing with circulating operational process of bone remodeling is a continuous physiological
monocytes with access to BRC255 has challenged this view. While entity. All events, from the formation of the bone remodeling
osteoclast apoptosis is a rare event requiring high energy compartment with canopy cells to the colonization of a reversal zone
expenditure for the removal of the apoptotic debris, osteoclast by reversal cells in the early period of bone remodeling, generate the
disassembly into smaller unresorbing cells, which can revert to environment necessary for the recruitment and expansion of the
functional osteoclasts when needed, is a more efficient process. osteoblast pool and subsequent refilling of resorption lacunae (Fig. 3).
Two observations support the latter view: first, primary osteoclasts
have been identified at the cutting cone and secondary osteoclasts
have been shown to establish a functional link with reversal cells THE OSTEOBLASTIC CELLS POOL: RECRUITMENT, EXPANSION
(Figs. 1 and 3) as focal bone remodeling progresses within a single AND OSTEOCYTOGENESIS
BRC,256 and second, osteoclasts have recently been shown to be Osteoblasts are cuboidal-shaped cells on bone surfaces. Osteo-
recycled into osteomorphs. Osteomorphs have been described as blasts have traditionally been considered the cell responsible for

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
13
the production of the organic components of the bone matrix and Upon the initiation of bone remodeling, as previously discussed,
its consequent mineralization because they synthetize and secrete the formation of BRCs by BLCs/canopy cells establishes a direct
matrix components such as collagen type I, OCN, osteonectin, connection with the vascular sinus closely covering bone-forming
bone sialoprotein, OPN, proteoglycans, and ALP.267 In vivo, a set of surfaces, allowing pericyte detachment and differentiation toward
osteoblasts, all with similar morphologies, lay down osteoid on the osteogenic phenotype.
active bone-forming surfaces.268 The dense endoplasmic reticu- Cells with true osteogenic potential can also circulate.278 Cells
lum of these cells indicates high synthetic activity, and the with multiple potential differentiation routes similar to those of
extensive contacts among these osteoblasts, osteocytes and BLCs BMSCs can enter the circulation, and although the mechanism by
indicate their extensive intercellular connections.269 which they enter the circulation is unclear, they circulate in
Marotti et al.270 hypothesized that in human bone physiology, physiologically significant numbers, correlate with markers of
osteoblasts can participate in two different types of osteogenesis: bone formation and increase during bone growth.127 Character-
static and dynamic osteogenesis. Static osteogenesis occurs in the ization of these cells deserves further study, and circulating
mesenchymal tissue to initiate intramembranous ossification to skeletal progenitors might form an additional pool of osteoblast
generate primary trabecula during growth. This osteogenesis type precursors for bone remodeling and/or tissue repair. In fact,
is thus independent of the loading environment and will be fractures result in the mobilization of mesenchymal osteogenic
subject to later osteoclast resorption. Static osteogenesis involves progenitors into the circulation to promote fracture healing, even
stationary osteoblasts, which are pluristratified and polarized in all in very old patients.279 Although circulating osteoprogenitor fate
directions. These osteoblasts differentiate into osteocytes in the and function are determined by the homing of these cells to focal
exact same site where they had been originated from the sites and their exposure to microenvironment signaling,280 their
mesenchymal precursors.271 Dynamic osteogenesis takes place to presence per sé in the blood has important implications for
thicken the primitive trabeculae generated in the context of static regenerative medicine.281
osteogenesis, and it is therefore the type of osteogenesis activated The average lifespan of active osteoblasts is 3 months.4 When
in response to metabolic and mechanical demand. The osteo- bone matrix formation ends and the subsequent matrix miner-
blasts participating in dynamic osteogenesis are arranged in an alization process completes the primary phase, mature osteoblasts
epithelial-like manner within a single cell layer. They all are undergo three possible fates. Between 60% and 80% of
polarized in the same direction, and they maintain contacts with osteoblasts at resorption lacunae die via apoptosis.282 Cells at all
osteocytic dendrites as they move from the mineralization front differentiation stages undergo apoptosis, which continues
toward the vascular surface.271 In the adult skeleton, physiological throughout the osteoblast life span and is regulated by Wnt
bone remodeling involves only dynamic osteogenesis, which refill signaling, PTH and mechanical stimulation.61 Osteoblast apoptosis
resorption lacunae. is key to regulating the extent and duration of bone formation and
Osteoblastic cells contribute not only to osteogenesis but also is the target of catabolic and anabolic regulatory factors affecting
to osteoclastogenesis by producing RANKL. The same osteo- bone mass. Apoptosis exerts a significant effect on the number of
genic cells may accomplish this dual function at different stages osteoblasts producing bone matrix at a bone formation site.283
of maturation. Immature osteoblasts respond to bone regulatory The balance between the expression of proapoptotic factors Bim
factors in a pro-osteoclastogenic manner, but during the and Bak and the prosurvival factor Bcl determines the osteoblast
maturation process, they lose RANKL expression and do not life span and, therefore, bone volume and strength.284 Estrogens
support osteoclastogenesis.272 Moreover, as cells mature, they and androgens also exert effects on bone cell life span. In
secrete more ECM and exert local inhibitory control over particular, they exert antiapoptotic effects on osteoblasts and
osteoclastogenesis by releasing the osteoclastogenic inhibitor osteocytes and proapoptotic effects on osteoclasts. Interestingly,
OPG.77 Osteoblasts are considered the major sources of OPG in estrogen receptors or the androgen receptor can transmit
cancellous bone, acting in concert with osteocytes and other antiapoptotic signals with similar efficiency, regardless of whether
cells to modulate the RANKL/RANK ratio and reduce resorption the ligand is an estrogen or an androgen.285 p53, a potent tumor
by osteoclasts. This inhibitory activity is mandatory for the suppressor, is also critically involved in determining osteoblast life
correct shift from bone resorption to bone formation and span, as it induces apoptosis, inhibits osteoblast proliferation and
protects newly formed bone against untimely resorption.77 stimulates osteoblast differentiation through the Akt–FoxOs
Cawley’s study stressed the concept “that the local concentration pathway286 to regulate bone remodeling.287
of OPG near the cell surface is a key factor in its ability to block the The remaining osteoblasts (those on the bone trabecular
activity of RANKL”. endosteal and endocortical surfaces) might differentiate into BLCs,
The osteoblast lineage derives from the differentiation of which are characterized by lower biosynthetic activity and a flat
mesenchymal cells in the stromal compartment of bone marrow morphology, or they might remain embedded in the matrix, undergo
(BM). The bone marrow stroma contains self-renewing, multi- the active process of osteocytogenesis,288 and become osteocytes
potent progenitors that can give rise to osteoblasts, thus ensuring (5%–20% of mature osteoblasts). Osteocytogenesis requires proteo-
a reservoir of bone-forming cells for bone growth, modeling and lytic activity to deepen the osteoid. Collagenase-resistant type I
remodeling.273 Anatomically, the BM stroma is part of the skeleton collagen in the bone matrix increased the osteocyte apoptosis
and is present in all bones except auditory ossicles, which rate.289 Additionally, the deletion of the metalloproteinase MT-MMP1
therefore do not undergo remodeling.274 The definition of skeletal generated an altered osteocytic network in mice.290 Osteocytogen-
stem cells and their physiological significance and terminology esis requires cytoskeleton plasticity to form dendrites (dendrogen-
associated with them have been debated.273–275 A self-renewing esis). Although the entire dendrogenesis process has not yet been
stromal progenitor, originally referred to as an “osteogenic” or elucidated, one factor known to be necessary in the dendrogenesis
“stromal”/”stem” cell (BMSC), corresponds to a specific type of of osteoid osteocytes (the late osteoblast/early osteocyte that
perivascular cell in mammalian bone marrow. The characterization undergoes the transition to mature osteocytes288) is E11/gp38 or
of pericytes as multipotent progenitors is evolving. Nevertheless, podoplanin (Pdpn), a transmembrane glycoprotein. The conditional
Sacchetti et al.276 pointed out that only BM vessel-residing cells deletion of PDNP in mice generated a decreased number and length
defined as pericytes have the ability to differentiate into of osteocyte processes.291 Recently, it has been shown that SP7
osteoblasts during physiological bone remodeling. Since tissue- (Osterix) is also necessary to support dendrogenesis because it
specific mesodermal progenitors show tissue-specific commit- regulates Osteocrine.292
ments in vivo, the native skeletogenic potential is thus intrinsic to With the progression of osteocytogenesis, osteoid osteocytes
the progenitor/stem cell set found in the skeleton.277 start expressing specific cytoskeletal markers (fimbrin and destrin)

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
14
and factors for mineralizing the surrounding matrix (Phex and 11. Manolagas, S. C. Birth and death of bone cells: basic regulatory mechanisms and
MEPE) until they become mineralizing osteocytes (expressing implications for the pathogenesis and treatment of osteoporosis. Endocr. Rev.
Dmp1). The cell terminally differentiate into mature osteocytes 21, 115–137 (2000).
producing SOST and FGF23.288 12. Jilka, R. L. Biology of the basic multicellular unit and the pathophysiology of
osteoporosis. Med. Pediatr. Oncol. 41, 182–185 (2003).
The differentiation of osteoblasts into osteocytes at the end of
13. Parfitt, A. M. Misconceptions (2): turnover is always higher in cancellous than in
the bone remodeling process is essential to reestablish mechan- cortical bone. Bone 30, 807–809 (2002).
ical competence. Therefore, 9.1 million osteocytes are replaced 14. Bonewald, L. F. & Marcus, D. F. R. In Osteoporosis 3rd edn (eds Nelson, D. &
daily during bone remodeling.16 Hence, osteocytes can be Rosen, C.) 170–189 (Elsevier, 2008).
considered dynamic cells capable of reconstituted mechanical 15. Metz, L. N., Martin, R. B. & Turner, A. S. Histomorphometric analysis of the effects
sensitivity after replacement293 as well as the inhibitory control of osteocyte density on osteonal morphology and remodeling. Bone 33,
over osteoclastogenesis via the local production of OPG.77 753–759 (2003).
16. Buenzli, P. R. & Sims, N. A. Quantifying the osteocyte network in the human
skeleton. Bone 75, 144–150 (2015).
17. Marotti, G. & Palumbo, C. The mechanism of transduction of mechanical strains
CONCLUSIONS: BONE REMODELING AS A CONTINUOUS FLOW
into biological signals at the bone cellular level. Eur. J. Histochem. 51, 15–19
OF INFORMATION AND CONNECTED EVENTS (2007).
Current evidence on bone remodeling has indicated the complex- 18. Tiede-Lewis, L. M. et al. Degeneration of the osteocyte network in the C57BL/6
ity of this process by showing how events and effectors are mouse model of aging. Aging 9, 2190–2208 (2017).
connected and coordinated to ensure the removal of old bone 19. Lecanda, F. et al. Connexin43 deficiency causes delayed ossification, cra-
and maintain bone mechanical competence. Recent findings have niofacial abnormalities, and osteoblast dysfunction. J. Cell Biol. 151, 931–944
shown that bone remodeling is a physiological process consisting (2000).
of continuous operations with fast reactive components that 20. Chung, D. J. et al. Low peak bone mass and attenuated anabolic response to
confer survival advantage. After BRC formation, local coordinating parathyroid hormone in mice with an osteoblast-specific deletion of con-
nexin43. J. Cell Sci. 119, 4187–4198 (2006).
factors and events, acting in concert in time and space and
21. Zhang, Y. et al. Enhanced osteoclastic resorption and responsiveness to
responding to metabolic and mechanical regulators, affect the mechanical load in gap junction deficient bone. PLoS One 6, e23516 (2011).
final quantum of BMU activity. This complete picture overwrites 22. Green, J. Role of bone in regulation of systemic acid-base balance. Miner. Elec-
the classical view of bone remodeling organized in discrete stages. trolyte Metab. 20, 7–30 (1994).
The current knowledge indicates that each BMU component is 23. Bushinsky, D. A., Chabala, J. M. & Levi-Setti, R. Ion microprobe analysis of mouse
critical for different mechanisms depending on the specific calvariae in vitro: evidence for a “bone membrane”. Am. J. Physiol. Endocrinol.
differentiation stage of the cell. All the signals discussed in the Metab. 256, E152–E158 (1989).
present review affect the remodeling process. The outcome 24. Bushinsky, D. A., Gavrilov, K., Chabala, J. M., Featherstone, J. D. & Levi-Setti, R.
depends on the cells generating the stimulus, and the way in Effect of metabolic acidosis on the potassium content of bone. J. Bone Miner.
Res. 12, 1664–1671 (1997).
which the stimulus is transferred. Understanding how the events
25. Rubinacci, A., Benelli, F. D., Borgo, E. & Villa, I. Bone as an ion exchange system:
determining the focal balance between bone resorption and evidence for a pump-leak mechanism devoted to the maintenance of high
formation unfold within each BMU provides necessary biological bone K+. Am. J. Physiol. Endocrinol. Metab. 278, E15–E24 (2000).
background information that will help to better identify targets of 26. Dedic, C. et al. Calcium fluxes at the bone/plasma interface: acute effects of
therapeutic approaches to osteoporosis. parathyroid hormone (PTH) and targeted deletion of PTH/PTH-related peptide
(PTHrP) receptor in the osteocytes. Bone 116, 135–143 (2018).
27. Rubinacci, A. et al. Bone as an ion exchange system: evidence for a link between
ACKNOWLEDGEMENTS mechanotransduction and metabolic needs. Am. J. Physiol. Endocrinol. Metab.
We thank Springer Nature Author Services for providing highly professional editing. 282, E851–E864 (2002).
28. Qin, L., Liu, W., Cao, H. & Xiao, G. Molecular mechanosensors in osteocytes. Bone
Res. 8, 23 (2020).
ADDITIONAL INFORMATION 29. McNamara, L. M., Majeska, R. J., Weinbaum, S., Friedrich, V. & Schaffler, M. B.
Attachment of osteocyte cell processes to the bone matrix. Anat. Rec. 292,
Competing interests: The authors declare no competing interests.
355–363 (2009).
30. Cabahug-Zuckerman, P. et al. Potential role for a specialized β(3) integrin-based
structure on osteocyte processes in bone mechanosensation. J. Orthop. Res. 36,
REFERENCES 642–652 (2018).
1. Frost, H. M. Bone remodeling dynamics (Charles C Thomas Company, 1963). 31. Malone, A. M. D. et al. Primary cilia mediate mechanosensing in bone cells by a
2. Delaisse, J. M. et al. Re-thinking the bone remodeling cycle mechanism and the calcium-independent mechanism. Proc. Natl. Acad. Sci. USA. 104, 13325–13330
origin of bone loss. Bone 141, 115628 (2020). (2007).
3. Maggiano, C. M., Maggiano, I. S., Tiesler, V. G., Chi-Keb, J. R. & Stout, S. D. 32. Plotkin, L. I., Speacht, T. L. & Donahue, H. J. Cx43 and mechanotransduction in
Methods and theory in bone modeling drift: comparing spatial analyses of bone. Curr. Osteoporos. Rep. 13, 67–72 (2015).
primary bone distributions in the human humerus. J. Anat. 228, 190–202 (2016). 33. Wang, L. et al. Mechanical sensing protein PIEZO1 regulates bone homeostasis
4. Parfitt, A. M. Osteonal and hemi-osteonal remodeling: the spatial and temporal via osteoblast-osteoclast crosstalk. Nat. Commun. 11, 282 (2020).
framework for signal traffic in adult human bone. J. Cell. Biochem. 55, 273–286 34. Lyons, J. S. et al. Microtubules tune mechanotransduction through NOX2 and
(1994). TRPV4 to decrease sclerostin abundance in osteocytes. Sci. Signal. 10, eaan5748
5. Frost, H. M. A synchronous group of mammalian cells whose in vivo behavior (2017).
can be studied. Henry Ford. Hosp. Med. Bull. 13, 161–172 (1965). 35. Morrell, A. E. et al. Mechanically induced Ca(2+) oscillations in osteocytes
6. Feynman, R., Leighton, R. & Sands, M. The Feynman lectures on physics (Addison- release extracellular vesicles and enhance bone formation. Bone Res. 6, 6
Wesley Pub. Co., 1964). (2018).
7. Parfitt, A. M. The cellular basis of bone remodeling: the quantum concept 36. Colombo, M., Raposo, G. & Théry, C. Biogenesis, secretion, and intercellular
reexamined in light of recent advances in the cell biology of bone. Calcif. Tissue interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev.
Int. 36, S37–S45 (1984). Biol. 30, 255–289 (2014).
8. Eriksen, E. F. Normal and pathological remodeling of human trabecular bone: 37. Kamel-ElSayed, S. A., Tiede-Lewis, L. M., Lu, Y., Veno, P. A. & Dallas, S. L. Novel
three dimensional reconstruction of the remodeling sequence in normals and in approaches for two and three dimensional multiplexed imaging of osteocytes.
metabolic bone disease. Endocr. Rev. 7, 379–408 (1986). Bone 76, 129–140 (2015).
9. Parfitt, A. M. The quantum concept of bone remodeling and turnover: impli- 38. Baron, R. & Kneissel, M. Wnt signaling in bone homeostasis and disease: from
cations for the pathogenesis of osteoporosis. Calcif. Tissue Int. 28, 1–5 (1979). human mutations to treatments. Nat. Med. 19, 179–192 (2013).
10. Marais, A. et al. The future of quantum biology. J. R. Soc. Interface 15, 20180640 39. Karner, C. M. & Long, F. Wnt signaling and cellular metabolism in osteoblasts.
(2018). Cell. Mol. Life Sci. 74, 1649–1657 (2017).

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
15
40. Day, T. F., Guo, X., Garrett-Beal, L. & Yang, Y. Wnt/β-catenin signaling in 70. Verborgt, O., Tatton, N. A., Majeska, R. J. & Schaffler, M. B. Spatial distribution of
mesenchymal progenitors controls osteoblast and chondrocyte differentiation bax and Bcl-2 in osteocytes after bone fatigue: complementary roles in bone
during vertebrate skeletogenesis. Dev. Cell 8, 739–750 (2005). remodeling regulation? J. Bone Miner. Res. 17, 907–914 (2002).
41. Hu, H. et al. Sequential roles of Hedgehog and Wnt signaling in osteoblast 71. Takeuchi, O. & Akira, S. Pattern recognition receptors and inflammation. Cell
development. Development 132, 49–60 (2005). 140, 805–820 (2010).
42. Song, L. et al. Loss of Wnt/β-catenin signaling causes cell fate shift of pre- 72. Messmer, D. et al. High mobility group box protein 1: an endogenous signal for
osteoblasts from osteoblasts to adipocytes. J. Bone Miner. Res. 27, 2344–2358 dendritic cell maturation and Th1 polarization. J. Immunol. 173, 307–313 (2004).
(2012). 73. Mori, T. et al. IL-1β and TNFα-initiated IL-6-STAT3 pathway is critical in mediating
43. Gaur, T. et al. Canonical Wnt signaling promotes osteogenesis by directly sti- inflammatory cytokines and RANKL expression in inflammatory arthritis. Int.
mulating Runx2 gene expression. J. Biol. Chem. 280, 33132–33140 (2005). Immunol. 23, 701–712 (2011).
44. Sato, T. et al. A FAK/HDAC5 signaling axis controls osteocyte mechan- 74. Andreev, D. et al. Osteocyte necrosis triggers osteoclast-mediated bone loss
otransduction. Nat. Commun. 11, 3282 (2020). through macrophage-inducible C-type lectin. J. Clin. Investig. 130, 4811–4830
45. Wein, M. N. et al. HDAC5 controls MEF2C-driven sclerostin expression in (2020).
osteocytes. J. Bone Miner. Res. 30, 400–411 (2015). 75. Glass, D. A. et al. Canonical Wnt signaling in differentiated osteoblasts controls
46. Wein, M. N. et al. SIKs control osteocyte responses to parathyroid hormone. Nat. osteoclast differentiation. Dev. Cell 8, 751–764 (2005).
Commun. 7, 13176 (2016). 76. Walsh, M. C. & Choi, Y. Biology of the RANKL-RANK-OPG system in immunity,
47. Stegen, S. et al. Osteocytic oxygen sensing controls bone mass through epi- bone, and beyond. Front. Immunol. 5, 511 (2014).
genetic regulation of sclerostin. Nat. Commun. 9, 2557 (2018). 77. Cawley, K. M. et al. Local production of osteoprotegerin by osteoblasts sup-
48. Verborgt, O., Gibson, G. J. & Schaffler, M. B. Loss of osteocyte integrity in presses bone resorption. Cell Rep. 32, 108052 (2020).
association with microdamage and bone remodeling after fatigue in vivo. J. 78. Delgado-Calle, J. et al. Control of bone anabolism in response to mechanical
Bone Miner. Res. 15, 60–67 (2000). loading and PTH by distinct mechanisms downstream of the PTH receptor. J.
49. Boyce, B. F. & Xing, L. Functions of RANKL/RANK/OPG in bone modeling and Bone Miner. Res. 32, 522–535 (2017).
remodeling. Arch. Biochem. Biophys. 473, 139–146 (2008). 79. Ben-Awadh, A. N. et al. Parathyroid hormone receptor signaling induces bone
50. Holliday, L. S., Patel, S. S. & Rody, W. J. Jr. RANKL and RANK in extracellular resorption in the adult skeleton by directly regulating the RANKL gene in
vesicles: surprising new players in bone remodeling. Extracell. Vesicles Circ. osteocytes. Endocrinology 155, 2797–2809 (2014).
Nucleic Acids 2, 18–28 (2021). 80. Rhee, Y. et al. PTH receptor signaling in osteocytes governs periosteal bone
51. Nakashima, T. et al. Evidence for osteocyte regulation of bone homeostasis formation and intracortical remodeling. J. Bone Miner. Res. 26, 1035–1046 (2011).
through RANKL expression. Nat. Med. 17, 1231–1234 (2011). 81. Saini, V. et al. Parathyroid hormone (PTH)/PTH-related peptide type 1 receptor
52. Xiong, J. et al. Matrix-embedded cells control osteoclast formation. Nat. Med. 17, (PPR) signaling in osteocytes regulates anabolic and catabolic skeletal responses
1235–1241 (2011). to PTH. J. Biol. Chem. 288, 20122–20134 (2013).
53. Lim, J., Burclaff, J., He, G., Mills, J. C. & Long, F. Unintended targeting of Dmp1- 82. Hongo, H. et al. Osteocytic osteolysis in PTH-treated wild-type and rankl(-/-)
Cre reveals a critical role for Bmpr1a signaling in the gastrointestinal mice examined by transmission electron microscopy, atomic force microscopy,
mesenchyme of adult mice. Bone Res. 5, 16049 (2017). and isotope microscopy. J. Histochem. Cytochem. 68, 651–668 (2020).
54. Xiong, J. et al. Osteocytes, not osteoblasts or lining cells, are the main source of 83. Tazawa, K. et al. Osteocytic osteolysis observed in rats to which parathyroid
the RANKL required for osteoclast formation in remodeling bone. PLoS One 10, hormone was continuously administered. J. Bone Miner. Metab. 22, 524–529
e0138189 (2015). (2004).
55. Zhao, S. et al. MLO-Y4 osteocyte-like cells support osteoclast formation and 84. Misof, B. M. et al. Bone matrix mineralization and osteocyte lacunae char-
activation. J. Bone Miner. Res. 17, 2068–2079 (2002). acteristics in patients with chronic kidney disease—mineral bone disorder(CKD-
56. Al-Dujaili, S. A. et al. Apoptotic osteocytes regulate osteoclast precursor MBD). J. Musculoskelet. Neuronal Interact. 19, 196–206 (2019).
recruitment and differentiation in vitro. J. Cell. Biochem. 112, 2412–2423 85. Gardinier, J. D., Al-Omaishi, S., Morris, M. D. & Kohn, D. H. PTH signaling mediates
(2011). perilacunar remodeling during exercise. Matrix Biol. 52-54, 162–175 (2016).
57. Honma, M. et al. RANKL subcellular trafficking and regulatory mechanisms in 86. Qing, H. et al. Demonstration of osteocytic perilacunar/canalicular remodeling in
osteocytes. J. Bone Miner. Res. 28, 1936–1949 (2013). mice during lactation. J. Bone Miner. Res. 27, 1018–1029 (2012).
58. Honma, M., Ikebuchi, Y. & Suzuki, H. Mechanisms of RANKL delivery to the 87. Misof, B. M. et al. No role of osteocytic osteolysis in the development and
osteoclast precursor cell surface. J. Bone Miner. Metab. 39, 27–33 (2021). recovery of the bone phenotype induced by severe secondary hyperpar-
59. Xiong, J. et al. Soluble RANKL contributes to osteoclast formation in adult mice athyroidism in vitamin D receptor deficient mice. Int. J. Mol. Sci. 21, 7989
but not ovariectomy-induced bone loss. Nat. Commun. 9, 2909 (2018). (2020).
60. Mizuno, A. et al. Transgenic mice overexpressing soluble osteoclast differ- 88. Davesne, D. et al. The phylogenetic origin and evolution of acellular bone in
entiation factor (sODF) exhibit severe osteoporosis. J. Bone Miner. Metab. 20, teleost fishes: insights into osteocyte function in bone metabolism. Biol. Rev.
337–344 (2002). Camb. Philos. Soc. 94, 1338–1363 (2019).
61. Komori, T. Cell death in chondrocytes, osteoblasts, and osteocytes. Int. J. Mol. Sci. 89. Hung, J. T. et al. Assessing the ability of zebrafish scales to contribute to the
17, 2045 (2016). short-term homeostatic regulation of [Ca2+] in the extracellular fluid during
62. Emerton, K. B. et al. Osteocyte apoptosis and control of bone resorption fol- calcemic challenges. Fish. Sci. 85, 943–959 (2019).
lowing ovariectomy in mice. Bone 46, 577–583 (2010). 90. Marenzana, M., Shipley, A. M., Squitiero, P., Kunkel, J. G. & Rubinacci, A. Bone as
63. Kennedy, O. D., Laudier, D. M., Majeska, R. J., Sun, H. B. & Schaffler, M. B. an ion exchange organ: evidence for instantaneous cell-dependent calcium
Osteocyte apoptosis is required for production of osteoclastogenic signals fol- efflux from bone not due to resorption. Bone 37, 545–554 (2005).
lowing bone fatigue in vivo. Bone 64, 132–137 (2014). 91. Farr, J. N., Kaur, J., Doolittle, M. L. & Khosla, S. Osteocyte cellular senescence. Curr.
64. Aguirre, J. I. et al. Osteocyte apoptosis is induced by weightlessness in mice and Osteoporos. Rep. 18, 559–567 (2020).
precedes osteoclast recruitment and bone loss. J. Bone Miner. Res. 21, 605–615 92. Farr, J. N. et al. Identification of senescent cells in the bone microenvironment. J.
(2006). Bone Miner. Res. 31, 1920–1929 (2016).
65. Kennedy, O. D. et al. Activation of resorption in fatigue-loaded bone involves 93. Uda, Y. et al. Parathyroid hormone signaling in mature osteoblasts/osteocytes
both apoptosis and active pro-osteoclastogenic signaling by distinct osteocyte protects mice from age-related bone loss. Aging 13, 25607–25642 (2021).
populations. Bone 50, 1115–1122 (2012). 94. Hayashi, M. et al. Autoregulation of osteocyte Sema3A orchestrates estrogen
66. Kringelbach, T. M., Aslan, D., Novak, I., Schwarz, P. & Jørgensen, N. R. UTP- action and counteracts bone aging. Cell Metab. 29, 627–637.e5 (2019).
induced ATP release is a fine-tuned signalling pathway in osteocytes. Purinergic 95. Sharma, D. et al. The effects of estrogen deficiency on cortical bone micro-
Signal 10, 337–347 (2014). porosity and mineralization. Bone 110, 1–10 (2018).
67. Cheung, W. Y. et al. Pannexin-1 and P2X7-receptor are required for apoptotic 96. Jackson, E. et al. Osteocyte Wnt/β-catenin pathway activation upon mechanical
osteocytes in fatigued bone to trigger RANKL production in neighboring loading is altered in ovariectomized mice. Bone Rep. 15, 101129 (2021).
bystander osteocytes. J. Bone Miner. Res. 31, 890–899 (2016). 97. Ma, L. et al. Connexin 43 hemichannels protect bone loss during estrogen
68. Luckprom, P., Wongkhantee, S., Yongchaitrakul, T. & Pavasant, P. Adenosine deficiency. Bone Res. 7, 11 (2019).
triphosphate stimulates RANKL expression through P2Y1receptor-cyclo- 98. Dole, N. S. et al. Osteocyte-intrinsic TGF-β signaling regulates bone quality
oxygenase-dependent pathway in human periodontal ligament cells. J. Period- through perilacunar/canalicular remodeling. Cell Rep. 21, 2585–2596 (2017).
ontal. Res. 45, 404–411 (2010). 99. Turner, A. G., Hanrath, M. A., Morris, H. A., Atkins, G. J. & Anderson, P. H. The local
69. Elliott, M. R. et al. Nucleotides released by apoptotic cells act as a find-me signal production of 1,25(OH)2D3 promotes osteoblast and osteocyte maturation. J.
to promote phagocytic clearance. Nature 461, 282–286 (2009). Steroid Biochem. Mol. Biol. 144, 114–118 (2014).

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
16
100. Yashiro, M. et al. Active vitamin D and vitamin D analogs stimulate fibroblast 132. Qiu, H., Xiao, W., Yue, J. & Wang, L. Heparan sulfate modulates Slit3-induced
growth factor 23 production in osteocyte-like cells via the vitamin D receptor. J. endothelial cell migration. Methods Mol. Biol. 1229, 549–555 (2015).
Pharm. Biomed. Anal. 182, 113139 (2020). 133. Geutskens, S. B. et al. Control of human hematopoietic stem/progenitor cell
101. Nociti, F. H. Jr. et al. Vitamin D represses dentin matrix protein 1 in cemento- migration by the extracellular matrix protein Slit3. Lab. Investig. 92, 1129–1139
blasts and osteocytes. J. Dent. Res. 93, 148–154 (2014). (2012).
102. Rolvien, T. et al. Vitamin D regulates osteocyte survival and perilacunar remo- 134. Zhou, B. O., Yue, R., Murphy, M. M., Peyer, J. G. & Morrison, S. J. Leptin-receptor-
deling in human and murine bone. Bone 103, 78–87 (2017). expressing mesenchymal stromal cells represent the main source of bone
103. Yuan, Y. et al. Impaired 1,25 dihydroxyvitamin D3 action and hypopho- formed by adult bone marrow. Cell Stem Cell 15, 154–168 (2014).
sphatemia underlie the altered lacuno-canalicular remodeling observed in the 135. Caire, R. et al. Parathyroid hormone remodels bone transitional vessels and the
Hyp mouse model of XLH. PLoS One 16, e0252348 (2021). leptin receptor‐positive pericyte network in mice. J. Bone Miner. Res. 34,
104. Ito, N., Findlay, D. M., Anderson, P. H., Bonewald, L. F. & Atkins, G. J. Extracellular 1487–1501 (2019).
phosphate modulates the effect of 1α,25-dihydroxy vitamin D3 (1,25D) on 136. Ominsky, M. S. et al. Differential temporal effects of sclerostin antibody and
osteocyte like cells. J. Steroid Biochem. Mol. Biol. 136, 183–186 (2013). parathyroid hormone on cancellous and cortical bone and quantitative differ-
105. Takashi, Y. & Fukumoto, S. Phosphate-sensing and regulatory mechanism of ences in effects on the osteoblast lineage in young intact rats. Bone 81, 380–391
FGF23 production. J. Endocrinol. Investig. 43, 877–883 (2020). (2015).
106. Gooi, J. H. et al. Decline in calcitonin receptor expression in osteocytes with age. 137. Matic, I. et al. Quiescent bone lining cells are a major source of osteoblasts
J. Endocrinol. 221, 181–191 (2014). during adulthood. Stem Cells 34, 2930–2942 (2016).
107. Davey, R. A. & Findlay, D. M. Calcitonin: physiology or fantasy? J. Bone Miner. Res. 138. Collin-Osdoby, P. et al. Receptor activator of NF-kappa B and osteoprotegerin
28, 973–979 (2013). expression by human microvascular endothelial cells, regulation by inflamma-
108. He, Z. et al. Irisin inhibits osteocyte apoptosis by activating the Erk signaling tory cytokines, and role in human osteoclastogenesis. J. Biol. Chem. 276,
pathway in vitro and attenuates ALCT-induced osteoarthritis in mice. Bone 141, 20659–20672 (2001).
115573 (2020). 139. Søe, K., Delaisse, J. M. & Borggaard, X. G. Osteoclast formation at the bone
109. Wood, C. L., Pajevic, P. D. & Gooi, J. H. Osteocyte secreted factors inhibit skeletal marrow/bone surface interface: importance of structural elements, matrix, and
muscle differentiation. Bone Rep. 6, 74–80 (2017). intercellular communication. Semin. Cell Dev. Biol. 112, 8–15 (2020).
110. Du, J. H. et al. The function of Wnt ligands on osteocyte and bone remodeling. J. 140. Ferrier, J., Xia, S. L., Lagan, E., Aubin, J. E. & Heersche, J. N. M. Displacement and
Dent. Res. 98, 930–938 (2019). translocation of osteoblast-like cells by osteoclasts. J. Bone Miner. Res. 9,
111. Miller, S. C., Bowman, B. M., Smith, J. M. & Jee, W. S. S. Characterization of 1397–1405 (1994).
endosteal bone-lining cells from fatty marrow bone sites in adult beagles. Anat. 141. Karsdal, M. A., Fjording, M. S., Foged, N. T., Delaissé, J. M. & Lochter, A. Trans-
Rec. 198, 163–173 (1980). forming growth factor-β-induced osteoblast elongation regulates osteoclastic
112. Miller, S. C., de Saint-Georges, L., Bowman, B. M. & Jee, W. S. Bone lining cells: bone resorption through a p38 mitogen-activated protein kinase- and matrix
structure and function. Scanning Microsc. 3, 953–960 (1989). metalloproteinase-dependent pathway. J. Biol. Chem. 276, 39350–39358 (2001).
113. Everts, V. et al. The bone lining cell: its role in cleaning howship’s lacunae and 142. Saltel, F. et al. Transmigration: a new property of mature multinucleated
initiating bone formation. J. Bone Miner. Res. 17, 77–90 (2002). osteoclasts. J. Bone Miner. Res. 21, 1913–1923 (2006).
114. Bloemen, V., de Vries, T. J., Schoenmaker, T. & Everts, V. Intercellular adhesion 143. Perez-Amodio, S., Beertsen, W. & Everts, V. (Pre-)osteoclasts induce retraction of
molecule-1 clusters during osteoclastogenesis. Biochem. Biophys. Res. Commun. osteoblasts before their fusion to osteoclasts. J. Bone Miner. Res. 19, 1722–1731
385, 640–645 (2009). (2004).
115. Talmage, R. V. Morphological and physiological considerations in a new concept 144. Kajita, M. et al. Membrane-type 1 matrix metalloproteinase cleaves CD44 and
of calcium transport in bone. Am. J. Anat. 129, 467–476 (1970). promotes cell migration. J. Cell Biol. 153, 893–904 (2001).
116. Wongdee, K., Riengrojpitak, S., Krishnamra, N. & Charoenphandhu, N. Claudin 145. Blavier, L. & Delaisse, J. M. Matrix metalloproteinases are obligatory for the
expression in the bone-lining cells of female rats exposed to long-standing migration of preosteoclasts to the developing marrow cavity of primitive long
acidemia. Exp. Mol. Pathol. 88, 305–310 (2010). bones. J. Cell Sci. 108, 3649–3659 (1995).
117. Andersen, T. L. et al. A physical mechanism for coupling bone resorption and 146. Sato, T., Foged, N. T. & Delaissé, J. M. The migration of purified osteoclasts
formation in adult human bone. Am. J. Pathol. 174, 239–247 (2009). through collagen is inhibited by matrix metalloproteinase inhibitors. J. Bone
118. Fields, G. B. Interstitial collagen catabolism. J. Biol. Chem. 288, 8785–8793 (2013). Miner. Res. 13, 59–66 (1998).
119. Sprangers, S. & Everts, V. Molecular pathways of cell-mediated degradation of 147. Andersen, T. L. et al. A scrutiny of matrix metalloproteinases in osteoclasts:
fibrillar collagen. Matrix Biol. 75-76, 190–200 (2019). evidence for heterogeneity and for the presence of MMPs synthesized by other
120. Delaissé, J. M. et al. Matrix metalloproteinases (MMP) and cathepsin K contribute cells. Bone 35, 1107–1119 (2004).
differently to osteoclastic activities. Microsc. Res. Tech. 61, 504–513 (2003). 148. Holmbeck, K. et al. MT1-MMP-deficient mice develop dwarfism, osteopenia,
121. Shi, J. et al. Membrane-type MMPs enable extracellular matrix permissiveness arthritis, and connective tissue disease due to inadequate collagen turnover. Cell
and mesenchymal cell proliferation during embryogenesis. Dev. Biol. 313, 99, 81–92 (1999).
196–209 (2008). 149. Sato, T. et al. Identification of the membrane-type matrix metalloproteinase
122. Parfitt, A. M. The bone remodeling compartment: a circulatory function for bone MT1-MMP in osteoclasts. J. Cell Sci. 110, 589–596 (1997).
lining cells. J. Bone Miner. Res. 16, 1583–1585 (2001). 150. Ohuchi, E. et al. Membrane type 1 matrix metalloproteinase digests interstitial
123. Hauge, E. M., Qvesel, D., Eriksen, E. F., Mosekilde, L. & Melsen, F. Cancellous bone collagens and other extracellular matrix macromolecules. J. Biol. Chem. 272,
remodeling occurs in specialized compartments lined by cells expressing 2446–2451 (1997).
osteoblastic markers. J. Bone Miner. Res. 16, 1575–1582 (2001). 151. Engsig, M. T. et al. Matrix metalloproteinase 9 and vascular endothelial growth
124. Kristensen, H. B., Andersen, T. L., Marcussen, N., Rolighed, L. & Delaisse, J. M. factor are essential for osteoclast recruitment into developing long bones. J. Cell
Increased presence of capillaries next to remodeling sites in adult human Biol. 151, 879–889 (2000).
cancellous bone. J. Bone Miner. Res. 28, 574–585 (2013). 152. Chellaiah, M. A. & Ma, T. Membrane localization of membrane type 1 matrix
125. Díaz-Flores, L. et al. Pericytes. Morphofunction, interactions and pathology in a metalloproteinase by CD44 regulates the activation of pro-matrix metallopro-
quiescent and activated mesenchymal cell niche. Histol. Histopathol. 24, teinase 9 in osteoclasts. Biomed. Res. Int. 2013, 302392 (2013).
909–969 (2009). 153. Zhu, L. et al. Osteoclast-mediated bone resorption is controlled by a compen-
126. Delaisse, J. M. The reversal phase of the bone-remodeling cycle: cellular pre- satory network of secreted and membrane-tethered metalloproteinases. Sci.
requisites for coupling resorption and formation. Bonekey Rep. 3, 561 (2014). Transl. Med. 12, eaaw6143 (2020).
127. Eghbali-Fatourechi, G. Z. et al. Circulating osteoblast-lineage cells in humans. N. 154. Møller, A. M. J. et al. Aging and menopause reprogram osteoclast precursors for
Engl. J. Med. 352, 1959–1966 (2005). aggressive bone resorption. Bone Res. 8, 27 (2020).
128. Kusumbe, A. P., Ramasamy, S. K. & Adams, R. H. Coupling of angiogenesis and 155. Weinstein, R. S., Roberson, P. K. & Manolagas, S. C. Giant osteoclast formation
osteogenesis by a specific vessel subtype in bone. Nature 507, 323–328 (2014). and long-term oral bisphosphonate therapy. N. Engl. J. Med. 360, 53–62 (2009).
129. Ramasamy, S. K., Kusumbe, A. P., Wang, L. & Adams, R. H. Endothelial Notch 156. Jobke, B., Milovanovic, P., Amling, M. & Busse, B. Bisphosphonate-osteoclasts:
activity promotes angiogenesis and osteogenesis in bone. Nature 507, 376–380 changes in osteoclast morphology and function induced by antiresorptive
(2014). nitrogen-containing bisphosphonate treatment in osteoporosis patients. Bone
130. Xie, H. et al. PDGF-BB secreted by preosteoclasts induces angiogenesis during 59, 37–43 (2014).
coupling with osteogenesis. Nat. Med. 20, 1270–1278 (2014). 157. Takagi, Y. et al. Effect of nitrogen-containing bisphosphonates on osteo-
131. Kim, B. J. et al. Osteoclast-secreted SLIT3 coordinates bone resorption and for- clasts and osteoclastogenesis: an ultrastructural study. Microscopy 70,
mation. J. Clin. Investig. 128, 1429–1441 (2018). 302–307 (2021).

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
17
158. Liu, H. et al. Histochemical evidence of zoledronate inhibiting c-src expression 189. Stenbeck, G. & Horton, M. A. Endocytic trafficking in actively resorbing osteo-
and interfering with CD44/OPN-mediated osteoclast adhesion in the tibiae of clasts. J. Cell Sci. 117, 827–836 (2004).
mice. J. Mol. Histol. 46, 313–323 (2015). 190. Helfrich, M. H., Nesbitt, S. A., Dorey, E. L. & Horton, M. A. Rat osteoclasts adhere
159. Yagi, M. et al. DC-STAMP is essential for cell-cell fusion in osteoclasts and For- to a wide range of rgd (arg-gly-asp) peptide-containing proteins, including the
eign body giant cells. J. Exp. Med. 202, 345–351 (2005). bone sialoproteins and fibronectin, via a β3 integrin. J. Bone Miner. Res. 7,
160. Takito, J. & Nakamura, M. Heterogeneity and actin cytoskeleton in osteoclast 335–343 (1992).
and macrophage multinucleation. Int. J. Mol. Sci. 21, 6629 (2020). 191. Faccio, R. et al. Activation of αvβ3 integrin on human osteoclast-like cells sti-
161. Streicher, C. et al. Estrogen regulates bone turnover by targeting RANKL mulates adhesion and migration in response to osteopontin. Biochem. Biophys.
expression in bone lining cells. Sci. Rep. 7, 6460 (2017). Res. Commun. 249, 522–525 (1998).
162. Takayanagi, H. Osteoimmunology: shared mechanisms and crosstalk between 192. Ross, F. P. et al. Interactions between the bone matrix proteins osteopontin and
the immune and bone systems. Nat. Rev. Immunol. 7, 292–304 (2007). bone sialoprotein and the osteoclast integrin alpha v beta 3 potentiate bone
163. Karin, M. & Greten, F. R. NF-κB: linking inflammation and immunity to cancer resorption. J. Biol. Chem. 268, 9901–9907 (1993).
development and progression. Nat. Rev. Immunol. 5, 749–759 (2005). 193. Faccio, R. et al. Localization and possible role of two different alpha v beta 3
164. Hayden, M. S., West, A. P. & Ghosh, S. NF-κB and the immune response. Onco- integrin conformations in resting and resorbing osteoclasts. J. Cell Sci. 115,
gene 25, 6758–6780 (2006). 2919–2929 (2002).
165. Franzoso, G. et al. Requirement for NF-kappaB in osteoclast and B-cell devel- 194. Duong, L. T., Lakkakorpi, P., Nakamura, I. & Rodan, G. A. Integrins and signaling in
opment. Genes Dev. 11, 3482–3496 (1997). osteoclast function. Matrix Biol. 19, 97–105 (2000).
166. Xing, L. et al. NF-κB p50 and p52 expression is not required for RANK-expressing 195. Xiang, B. et al. The osteoclasts attach to the bone surface where the extracellular
osteoclast progenitor formation but is essential for RANK- and cytokine- calcium concentration decreases. Cell Biochem. Biophys. 74, 553–558 (2016).
mediated osteoclastogenesis. J. Bone Miner. Res. 17, 1200–1210 (2002). 196. Sims, N. A. & Martin, T. J. Osteoclasts provide coupling signals to osteoblast
167. Asagiri, M. et al. Autoamplification of NFATc1 expression determines its essential lineage cells through multiple mechanisms. Annu. Rev. Physiol. 82, 507–529
role in bone homeostasis. J. Exp. Med. 202, 1261–1269 (2005). (2020).
168. Ishida, N. et al. Large scale gene expression analysis of osteoclastogenesis 197. Jiang, X. et al. Method development of efficient protein extraction in bone tissue
in vitro and elucidation of NFAT2 as a key regulator. J. Biol. Chem. 277, for proteome analysis. J. Proteome Res. 6, 2287–2294 (2007).
41147–41156 (2002). 198. Rubinacci, A. Expanding the functional spectrum of vitamin K in bone. Focus on:
169. Takayanagi, H. et al. Induction and activation of the transcription factor NFATc1 “vitamin K promotes mineralization, osteoblast to osteocyte transition, and an
(NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev. anti-catabolic phenotype by γ-carboxylation-dependent and -independent
Cell 3, 889–901 (2002). mechanisms”. Am. J. Physiol. Cell Physiol. 297, C1336–C1338 (2009).
170. Ruocco, M. G. et al. I{kappa}B kinase (IKK){beta}, but not IKK{alpha}, is a critical 199. Mizokami, A., Kawakubo-Yasukochi, T. & Hirata, M. Osteocalcin and its endocrine
mediator of osteoclast survival and is required for inflammation-induced bone functions. Biochem. Pharmacol. 132, 1–8 (2017).
loss. J. Exp. Med. 201, 1677–1687 (2005). 200. Lin, X., Patil, S., Gao, Y. G. & Qian, A. The bone extracellular matrix in bone
171. Xiu, Y. et al. Chloroquine reduces osteoclastogenesis in murine osteoporosis by formation and regeneration. Front. Pharmacol. 11, 757 (2020).
preventing TRAF3 degradation. J. Clin. Investig. 124, 297–310 (2014). 201. Hering, S. et al. TGFβ1 and TGFβ2 mRNA and protein expression in human bone
172. Luo, J. et al. LGR4 is a receptor for RANKL and negatively regulates osteoclast samples. Exp. Clin. Endocrinol. Diabetes 109, 217–226 (2001).
differentiation and bone resorption. Nat. Med. 22, 539–546 (2016). 202. Oursler, M. J. Osteoclast synthesis and secretion and activation of latent trans-
173. Ko, Y. J. et al. A novel modified RANKL variant can prevent osteoporosis by forming growth factor β. J. Bone Miner. Res. 9, 443–452 (1994).
acting as a vaccine and an inhibitor. Clin. Transl. Med. 11, e368 (2021). 203. Tang, Y. et al. TGF-beta1-induced migration of bone mesenchymal stem cells
174. Yang, X., Pande, S., Scott, C. & Friesel, R. Macrophage colony-stimulating factor couples bone resorption with formation. Nat. Med. 15, 757–765 (2009).
pretreatment of bone marrow progenitor cells regulates osteoclast differentia- 204. Alliston, T., Choy, L., Ducy, P., Karsenty, G. & Derynck, R. TGF-beta-induced
tion based upon the stage of myeloid development. J. Cell. Biochem. 120, repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and
12450–12460 (2019). inhibits osteoblast differentiation. EMBO J. 20, 2254–2272 (2001).
175. Faccio, R., Takeshita, S., Zallone, A., Ross, F. P. & Teitelbaum, S. L. c-Fms and the 205. Breen, E. C. et al. TGF? alters growth and differentiation related gene expression
αvβ3 integrin collaborate during osteoclast differentiation. J. Clin. Investig. 111, in proliferating osteoblasts in vitro, preventing development of the mature
749–758 (2003). bone phenotype. J. Cell. Physiol. 160, 323–335 (1994).
176. Scholtysek, C. et al. NR4A1 regulates motility of osteoclast precursors and serves 206. Weivoda, M. M. et al. Osteoclast TGF-β receptor signaling induces
as target for the modulation of systemic bone turnover. J. Bone Miner. Res. 33, Wnt1 secretion and couples bone resorption to bone formation. J. Bone Miner.
2035–2047 (2018). Res. 31, 76–85 (2016).
177. Koga, T. et al. Costimulatory signals mediated by the ITAM motif cooperate with 207. Li, J. et al. TGFβ-induced degradation of TRAF3 in mesenchymal progenitor cells
RANKL for bone homeostasis. Nature 428, 758–763 (2004). causes age-related osteoporosis. Nat. Commun. 10, 2795 (2019).
178. Souza, P. P. C. & Lerner, U. H. Finding a toll on the route: the fate of 208. Kim, J. S. et al. Transforming growth factor-β1 regulates macrophage migration
osteoclast progenitors after toll-like receptor activation. Front. Immunol. 10, via RhoA. Blood 108, 1821–1829 (2006).
1663 (2019). 209. Takai, H. et al. Transforming growth factor-β stimulates the production of
179. Yim, M. The role of toll-like receptors in osteoclastogenesis. J. Bone Metab. 27, osteoprotegerin/osteoclastogenesis inhibitory factor by bone marrow stromal
227–235 (2020). cells. J. Biol. Chem. 273, 27091–27096 (1998).
180. Maeda, K. et al. Wnt5a-Ror2 signaling between osteoblast-lineage cells and 210. Houde, N., Chamoux, E., Bisson, M. & Roux, S. Transforming growth factor-beta1
osteoclast precursors enhances osteoclastogenesis. Nat. Med. 18, 405–412 (TGF-beta1) induces human osteoclast apoptosis by up-regulating Bim. J. Biol.
(2012). Chem. 284, 23397–23404 (2009).
181. Kobayashi, Y. et al. Wnt16 regulates osteoclast differentiation in conjunction 211. Ota, K. et al. TGF-β induces Wnt10b in osteoclasts from female mice to enhance
with Wnt5a. Biochem. Biophys. Res. Commun. 463, 1278–1283 (2015). coupling to osteoblasts. Endocrinology 154, 3745–3752 (2013).
182. Uehara, S. et al. Protein kinase N3 promotes bone resorption by osteoclasts in 212. Seck, T. et al. Concentration of insulin-like growth factor (IGF)-I and -II in iliac
response to Wnt5a-Ror2 signaling. Sci. Signal. 10, eaan0023 (2017). crest bone matrix from pre- and postmenopausal women: relationship to age,
183. Okamoto, M. et al. Noncanonical Wnt5a enhances Wnt/β-catenin signaling menopause, bone turnover, bone volume, and circulating IGFs. J. Clin. Endo-
during osteoblastogenesis. Sci. Rep. 4, 4493 (2014). crinol. Metab. 83, 2331–2337 (1998).
184. Roberts, J. L. et al. Deletion of Wnt5a in osteoclasts results in bone loss through 213. Lean, J. M., Mackay, A. G., Chow, J. W. & Chambers, T. J. Osteocytic expression of
decreased bone formation. Ann. N. Y. Acad. Sci. 1463, 45–59 (2020). mRNA for c-fos and IGF-I: an immediate early gene response to an osteogenic
185. Akisaka, T. The ruffled border and attachment regions of the apposing mem- stimulus. Am. J. Physiol. Endocrinol. Metab. 270, E937–E945 (1996).
brane of resorbing osteoclasts as visualized from the cytoplasmic face of the 214. Lau, K. H. W. et al. Osteocyte-derived insulin-like growth factor I is essential for
membrane. J. Electron Microsc. 55, 53–61 (2006). determining bone mechanosensitivity. Am. J. Physiol. Endocrinol. Metab. 305,
186. Yao, G., Feng, H., Cai, Y., Qi, W. & Kong, K. Characterization of vacuolar-ATPase E271–E281 (2013).
and selective inhibition of vacuolar-H(+)-ATPase in osteoclasts. Biochem. Bio- 215. Fiedler, J., Brill, C., Blum, W. F. & Brenner, R. E. IGF-I and IGF-II stimulate directed
phys. Res. Commun. 357, 821–827 (2007). cell migration of bone-marrow-derived human mesenchymal progenitor cells.
187. Kornak, U. et al. Loss of the ClC-7 chloride channel leads to osteopetrosis in mice Biochem. Biophys. Res. Commun. 345, 1177–1183 (2006).
and man. Cell 104, 205–215 (2001). 216. Yuan, Y. et al. Gene expression profiles and bioinformatics analysis of insulin-like
188. Delaissé, J. M. et al. Proteinases in bone resorption: obvious and less obvious growth factor-1 promotion of osteogenic differentiation. Mol. Genet. Genom.
roles. Clin. Chim. Acta 291, 223–234 (2000). Med 7, e00921 (2019).

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
18
217. Xian, L. et al. Matrix IGF-1 maintains bone mass by activation of mTOR in 245. Amizuka, N. et al. Defective bone remodelling in osteoprotegerin-deficient mice.
mesenchymal stem cells. Nat. Med. 18, 1095–1101 (2012). J. Electron Microsc. 52, 503–513 (2003).
218. Liu, L. & Parent, C. A. Review series: TOR kinase complexes and cell migration. J. 246. Sone, E. et al. The induction of RANKL molecule clustering could stimulate early
Cell Biol. 194, 815–824 (2011). osteoblast differentiation. Biochem. Biophys. Res. Commun. 509, 435–440 (2019).
219. Feng, X. et al. Insulin-like growth factor 1 can promote proliferation and 247. Pitt, J. M., Kroemer, G. & Zitvogel, L. Extracellular vesicles: masters of intercellular
osteogenic differentiation of human dental pulp stem cells via mTOR pathway. communication and potential clinical interventions. J. Clin. Investig. 126,
Dev. Growth Differ. 56, 615–624 (2014). 1139–1143 (2016).
220. Sims, N. A. & Martin, T. J. Coupling signals between the osteoclast and osteo- 248. Ma, Q. et al. Osteoclast-derived apoptotic bodies show extended biological
blast: how are messages transmitted between these temporary visitors to the effects of parental cell in promoting bone defect healing. Theranostics 10,
bone surface? Front. Endocrinol. 6, 41 (2015). 6825–6838 (2020).
221. Karsdal, M. A., Neutzsky-Wulff, A. V., Dziegiel, M. H., Christiansen, C. & Henriksen, 249. Li, D. et al. Osteoclast-derived exosomal miR-214-3p inhibits osteoblastic bone
K. Osteoclasts secrete non-bone derived signals that induce bone formation. formation. Nat. Commun. 7, 10872 (2016).
Biochem. Biophys. Res. Commun. 366, 483–488 (2008). 250. Ma, Q. et al. Osteoclast-derived apoptotic bodies couple bone resorption and
222. Thudium, C. S. et al. A comparison of osteoclast-rich and osteoclast-poor formation in bone remodeling. Bone Res 9, 5 (2021).
osteopetrosis in adult mice sheds light on the role of the osteoclast in coupling 251. Zhao, C. et al. Bidirectional ephrinB2-EphB4 signaling controls bone home-
bone resorption and bone formation. Calcif. Tissue Int. 95, 83–93 (2014). ostasis. Cell Metab. 4, 111–121 (2006).
223. Neutzsky-Wulff, A. V. et al. Severe developmental bone phenotype in ClC-7 252. Tazaki, Y. et al. RANKL, Ephrin-Eph and Wnt10b are key intercellular commu-
deficient mice. Dev. Biol. 344, 1001–1010 (2010). nication molecules regulating bone remodeling in autologous transplanted
224. Henriksen, K. et al. Dissociation of bone resorption and bone formation in adult goldfish scales. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 225, 46–58 (2018).
mice with a non-functional V-ATPase in osteoclasts leads to increased bone 253. Matsuo, K. & Otaki, N. Bone cell interactions through Eph/ephrin: bone mod-
strength. PLoS One 6, e27482 (2011). eling, remodeling and associated diseases. Cell Adhes. Migr. 6, 148–156 (2012).
225. McClung, M. R. et al. Odanacatib for the treatment of postmenopausal osteo- 254. Allan, E. H. et al. EphrinB2 regulation by PTH and PTHrP revealed by molecular
porosis: results of the LOFT multicenter, randomised, double blind, placebo- profiling in differentiating osteoblasts. J. Bone Miner. Res. 23, 1170–1181 (2008).
controlled trial and LOFT Extension study. Lancet Diabetes Endocrinol. 7, 255. Jacome-Galarza, C. E. et al. Developmental origin, functional maintenance and
899–911 (2019). genetic rescue of osteoclasts. Nature 568, 541–545 (2019).
226. Guerrini, M. M. et al. Human osteoclast-poor osteopetrosis with hypo- 256. Lassen, N. E. et al. Coupling of bone resorption and formation in real time:
gammaglobulinemia due to TNFRSF11A (RANK) mutations. Am. J. Hum. Genet. new knowledge gained from human haversian BMUs. J. Bone Miner. Res. 32,
83, 64–76 (2008). 1395–1405 (2017).
227. Whyte, M. P. et al. Dysosteosclerosis presents as an “osteoclast-poor” form of 257. McDonald, M. M. et al. Osteoclasts recycle via osteomorphs during RANKL-
osteopetrosis: comprehensive investigation of a 3-year-old girl and literature stimulated bone resorption. Cell 184, 1330–1347.e13 (2021).
review. J. Bone Miner. Res. 25, 2527–2539 (2010). 258. Kim, S. W. et al. Intermittent parathyroid hormone administration converts
228. Walker, E. C. et al. Cardiotrophin-1 is an osteoclast-derived stimulus of bone quiescent lining cells to active osteoblasts. J. Bone Miner. Res. 27, 2075–2084
formation required for normal bone remodeling. J. Bone Miner. Res. 23, (2012).
2025–2032 (2008). 259. Makino, A. et al. Frequent administration of abaloparatide shows greater gains
229. Meshcheryakova, A., Mechtcheriakova, D. & Pietschmann, P. Sphingosine in bone anabolic window and bone mineral density in mice: a comparison with
1-phosphate signaling in bone remodeling: multifaceted roles and therapeutic teriparatide. Bone 142, 115651 (2021).
potential. Expert Opin. Ther. Targets 21, 725–737 (2017). 260. Zanchetta, M. B. et al. Significant bone loss after stopping long-term denosu-
230. Pederson, L., Ruan, M., Westendorf, J. J., Khosla, S. & Oursler, M. J. Regulation of mab treatment: a post FREEDOM study. Osteoporos. Int. 29, 41–47 (2018).
bone formation by osteoclasts involves Wnt/BMP signaling and the chemokine 261. Sølling, A. S., Harsløf, T. & Langdahl, B. Treatment with zoledronate subsequent
sphingosine-1-phosphate. Proc. Natl Acad. Sci. USA 105, 20764–20769 (2008). to denosumab in osteoporosis: a randomized trial. J. Bone Miner. Res. 35,
231. Takeshita, S. et al. Osteoclast-secreted CTHRC1 in the coupling of bone 1858–1870 (2020).
resorption to formation. J. Clin. Investig. 123, 3914–3924 (2013). 262. Baron, R., Vignery, A. & Van Tran, P. The significance of lacunar erosion without
232. Matsuoka, K., Park, K. A., Ito, M., Ikeda, K. & Takeshita, S. Osteoclast-derived osteoclasts: studies on the reversal phase of the remodeling sequence. Metab.
complement component 3a stimulates osteoblast differentiation. J. Bone Miner. Bone Dis. Relat. Res. 2, 35–40 (1980).
Res. 29, 1522–1530 (2014). 263. Dempster, D. W. et al. Standardized nomenclature, symbols, and units for bone
233. Cornish, J., Callon, K. E., Edgar, S. G. & Reid, I. R. Leukemia inhibitory factor is histomorphometry: a 2012 update of the report of the ASBMR histomorpho-
mitogenic to osteoblasts. Bone 21, 243–247 (1997). metry nomenclature committee. J. Bone Miner. Res. 28, 2–17 (2013).
234. Weivoda, M. M. et al. Identification of osteoclast-osteoblast coupling factors in 264. Yamamoto, T., Yamagata, A. & Nagai, H. A histochemical study of tartrate-
humans reveals links between bone and energy metabolism. Nat. Commun. 11, resistant acid phosphatase activity in rat osteoblasts. Acta Histochem. Cytochem.
87 (2020). 29, 221–225 (1996).
235. Kim, B. J. & Koh, J. M. Coupling factors involved in preserving bone balance. Cell. 265. Andersen, T. L. et al. Understanding coupling between bone resorption and
Mol. Life Sci. 76, 1243–1253 (2019). formation. Am. J. Pathol. 183, 1–12 (2013).
236. Liu, M., Sun, Y. & Zhang, Q. Emerging role of extracellular vesicles in bone 266. Abdelgawad, M. E. et al. Early reversal cells in adult human bone remodeling:
remodeling. J. Dent. Res. 97, 859–868 (2018). osteoblastic nature, catabolic functions and interactions with osteoclasts. His-
237. Huynh, N. et al. Characterization of regulatory extracellular vesicles from tochem. Cell Biol. 145, 603–615 (2016).
osteoclasts. J. Dent. Res. 95, 673–679 (2016). 267. Rosenberg, N., Rosenberg, O. & Soudry, M. Osteoblasts in bone physiology-mini
238. Ikebuchi, Y. et al. Coupling of bone resorption and formation by RANKL reverse review. Rambam Maimonides Med. J. 3, e0013 (2012).
signalling. Nature 561, 195–200 (2018). 268. Florencio-Silva, R., Sasso, G. R. D. S., Sasso-Cerri, E., Simões, M. J. & Cerri, P. S.
239. Furuya, Y. et al. Stimulation of bone formation in cortical bone of mice treated Biology of bone tissue: structure, function, and factors that influence bone cells.
with a receptor activator of nuclear factor-κB ligand (RANKL)-binding peptide Biomed. Res. Int. 2015, 421746 (2015).
that possesses osteoclastogenesis inhibitory activity. J. Biol. Chem. 288, 269. Yellowley, C. E., Li, Z., Zhou, Z., Jacobs, C. R. & Donahue, H. J. Functional gap
5562–5571 (2013). junctions between osteocytic and osteoblastic cells. J. Bone Miner. Res. 15,
240. Portal-Núñez, S. et al. Unexpected bone formation produced by RANKL block- 209–217 (2000).
ade. Trends Endocrinol. Metab. 28, 695–704 (2017). 270. Marotti, G., Ferretti, M., Palumbo, C. & Benincasa, M. Static anddynamic bone
241. Bone, H. G. et al. 10 years of denosumab treatment in postmenopausal women formation and the mechanism of collagen fiberorientation. Bone 25, 156 (1999).
with osteoporosis: results from the phase 3 randomised FREEDOM trial and 271. Ferretti, M. & Palumbo, C. The osteocyte: from “prisoner” to “orchestrator”. J.
open-label extension. Lancet Diabetes Endocrinol. 5, 513–523 (2017). Funct. Morphol. Kinesiol 6, 28 (2021).
242. Dempster, D. W. et al. Modeling‐based bone formation in the human femoral 272. Atkins, G. J. et al. RANKL expression is related to the differentiation state of
neck in subjects treated with denosumab. J. Bone Miner. Res. 35, 1282–1288 human osteoblasts. J. Bone Miner. Res. 18, 1088–1098 (2003).
(2020). 273. Bianco, P. & Robey, P. G. Skeletal stem cells. Development 142, 1023–1027
243. Wang, L., Huang, B., Chen, X. & Su, J. New insight into unexpected bone for- (2015).
mation by denosumab. Drug Discov. Today 25, 1919–1922 (2020). 274. Palumbo, C., Cavani, F., Sena, P., Benincasa, M. & Ferretti, M. Osteocyte apoptosis
244. Chen, X., Zhi, X., Wang, J. & Su, J. RANKL signaling in bone marrow mesenchymal and absence of bone remodeling in human auditory ossicles and scleral ossicles
stem cells negatively regulates osteoblastic bone formation. Bone Res 6, 34 of lower vertebrates: a mere coincidence or linked processes? Calcif. Tissue Int.
(2018). 90, 211–218 (2012).

Bone Research (2022)10:48


The operational process of bone remodeling
S Bolamperti et al.
19
275. Bianco, P. et al. The meaning, the sense and the significance: translating 287. Liu, H. & Li, B. p53 control of bone remodeling. J. Cell. Biochem. 111, 529–534
the science of mesenchymal stem cells into medicine. Nat. Med. 19, 35–42 (2010).
(2013). 288. Bonewald, L. F. The amazing osteocyte. J. Bone Miner. Res. 26, 229–238 (2011).
276. Sacchetti, B. et al. No identical “mesenchymal stem cells” at different times and 289. Zhao, W., Byrne, M. H., Wang, Y. & Krane, S. M. Osteocyte and osteoblast
sites: human committed progenitors of distinct origin and differentiation apoptosis and excessive bone deposition accompany failure of collagenase
potential are incorporated as adventitial cells in microvessels. Stem Cell Rep. 6, cleavage of collagen. J. Clin. Investig. 106, 941–949 (2000).
897–913 (2016). 290. Holmbeck, K. et al. The metalloproteinase MT1-MMP is required for normal
277. Sacchetti, B. Post-natal “mesenchymal” stem cells: the assayable skeletal development and maintenance of osteocyte processes in bone. J. Cell Sci. 118,
potency. J. Stem Cells Regen. Med. 15, 12–15 (2019). 147–156 (2005).
278. Kuznetsov, S. A. et al. Circulating skeletal stem cells. J. Cell Biol. 153, 1133–1140 291. Staines, K. A. et al. Hypomorphic conditional deletion of E11/podoplanin reveals
(2001). a role in osteocyte dendrite elongation. J. Cell. Physiol. 232, 3006–3019 (2017).
279. Alm, J. J. et al. Circulating plastic adherent mesenchymal stem cells in aged hip 292. Wang, J. S. et al. Control of osteocyte dendrite formation by Sp7 and its target
fracture patients. J. Orthop. Res. 28, 1634–1642 (2010). gene osteocrin. Nat. Commun. 12, 6271 (2021).
280. Canalis, E. The fate of circulating osteoblasts. N. Engl. J. Med 352, 2014–2016 293. Lerebours, C. & Buenzli, P. R. Towards a cell-based mechanostat theory of bone:
(2005). the need to account for osteocyte desensitisation and osteocyte replacement. J.
281. Feehan, J., Nurgali, K., Apostolopoulos, V., Al Saedi, A. & Duque, G. Circulating Biomech. 49, 2600–2606 (2016).
osteogenic precursor cells: building bone from blood. EBioMedicine 39, 603–611
(2019).
282. Manolagas, S. C. & Parfitt, A. M. What old means to bone. Trends Endocrinol. Open Access This article is licensed under a Creative Commons
Metab. 21, 369–374 (2010). Attribution 4.0 International License, which permits use, sharing,
283. Weinstein, R. S., Jilka, R. L., Parfitt, A. M. & Manolagas, S. C. Inhibition of osteo- adaptation, distribution and reproduction in any medium or format, as long as you give
blastogenesis and promotion of apoptosis of osteoblasts and osteocytes by appropriate credit to the original author(s) and the source, provide a link to the Creative
glucocorticoids. Potential mechanisms of their deleterious effects on bone. J. Commons license, and indicate if changes were made. The images or other third party
Clin. Investig. 102, 274–282 (1998). material in this article are included in the article’s Creative Commons license, unless
284. Moriishi, T. et al. Overexpression of BCLXL in osteoblasts inhibits osteoblast indicated otherwise in a credit line to the material. If material is not included in the
apoptosis and increases bone volume and strength. J. Bone Miner. Res. 31, article’s Creative Commons license and your intended use is not permitted by statutory
1366–1380 (2016). regulation or exceeds the permitted use, you will need to obtain permission directly
285. Manolagas, S. C., Kousteni, S. & Jilka, R. L. Sex steroids and bone. Recent Prog. from the copyright holder. To view a copy of this license, visit http://creativecommons.
Horm. Res. 57, 385–409 (2002). org/licenses/by/4.0/.
286. Wang, X. et al. p53 functions as a negative regulator of osteoblastogenesis,
osteoblast-dependent osteoclastogenesis, and bone remodeling. J. Cell Biol.
172, 115–125 (2006). © The Author(s) 2022

Bone Research (2022)10:48

You might also like